You are on page 1of 26

European Journal of Medicinal Chemistry 222 (2021) 113588

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research progress of dual inhibitors targeting crosstalk between


histone epigenetic modulators for cancer therapy
Ying-Chao Duan a, *, Shao-Jie Zhang a, Xiao-Jing Shi b, Lin-Feng Jin a, Tong Yu a, Yu Song a,
Yuan-Yuan Guan a, **
a
School of Pharmacy, Xinxiang Medical University, 453003, Xinxiang, Henan Province, PR China
b
Laboratory Animal Center, Academy of Medical Science, Zhengzhou University, 450052, Zhengzhou, Henan Province, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Abnormal epigenetics is a critical hallmark of human cancers. Anticancer drug discovery directed at
Received 27 March 2021 histone epigenetic modulators has gained impressive advances with six drugs available for cancer
Received in revised form therapy and numerous other candidates undergoing clinical trials. However, limited therapeutic profile,
9 May 2021
drug resistance, narrow safety margin, and dose-limiting toxicities pose intractable challenges for their
Accepted 25 May 2021
Available online 1 June 2021
clinical utility. Because histone epigenetic modulators undergo intricate crosstalk and act cooperatively
to shape an aberrant epigenetic profile, co-targeting histone epigenetic modulators with a different
mechanism of action has rapidly emerged as an attractive strategy to overcome the limitations faced by
Keywords:
Epigenetic modulator
the single-target epigenetic inhibitors. In this review, we summarize in detail the crosstalk of histone
Dual inhibitor epigenetic modulators in regulating gene transcription and the progress of dual epigenetic inhibitors
Anticancer therapy targeting this crosstalk.
Crosstalk © 2021 Elsevier Masson SAS. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Dual inhibitors targeting histone erasers and readers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Dual inhibitors targeting HDAC and BRD4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1.1. Crosstalk between HDAC and BRD4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1.2. Reported HDAC/BRD4 dual inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.2. Dual inhibitors targeting HDAC and BRPF1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
3. Dual inhibitors targeting histone erasers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
3.1. Dual inhibitors targeting LSD1 and HDAC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
3.1.1. Crosstalk between LSD1 and HDAC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
3.1.2. Reported LSD1/HDAC dual inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
3.2. Dual inhibitors targeting LSD1 and JmjC KDM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3.2.1. Crosstalk between LSD1 and JmjC KDM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3.2.2. Reported LSD1/JmjC KDM dual inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
4. Dual inhibitors targeting histone erasers and writers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
4.1. Dual inhibitors targeting HDAC and G9a . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
4.1.1. Crosstalk between HDAC and G9a . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
4.1.2. Reported HDAC/G9a dual inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4.2. Dual inhibitors targeting HDAC and EZH2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4.2.1. Crosstalk between HDAC and EZH2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

* Corresponding author.
** Corresponding author.
E-mail addresses: duanyingchao1986@163.com (Y.-C. Duan), 131011@xxmu.edu.
cn (Y.-Y. Guan).

https://doi.org/10.1016/j.ejmech.2021.113588
0223-5234/© 2021 Elsevier Masson SAS. All rights reserved.
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

4.2.2. Reported HDAC/EZH2 dual inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19


4.3. Dual inhibitors targeting LSD1 and G9a . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
5. Dual inhibitor targeting histone writer and reader . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5.1. Crosstalk between BRD4 and CBP/p300 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5.2. Reported BRD4-CBP/p300 dual inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
6. Discussion and conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Declaration of competing interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

1. Introduction in other words, specific residues may undergo multiple epigenetic


modifications, leading to distinct gene expression patterns. For
Epigenetic modifications of lysine-rich N-terminal histone example, lysine residues can be targeted for various modifications,
“tails” are primarily via acetylation and methylation, ubiquitina- such as acetylation, methylation, ubiquitination, or sumoylation
tion, phosphorylation, and sumoylation. These chemical modifica- and can harbor one, two, or three methylation marks. Moreover, a
tions can associate at distinct amino acid residues with significantly specific pattern of histone modifications frequently affects the
divergent transcriptional information. For example, H3K9 acetyla- generation or loss of other modifications, demonstrating extensive
tion and H3K4, H3K36, and H3K79 methylation are associated with connections, and crosstalk between histone modifications. For
open chromatin and gene transcription, whereas methylation of instance, acetylation on H3K4 or H3K9 and phosphorylated H3S10
H3K9, H3K27, and H4K20 suppress gene expression [1]. These or H3T11 can block the G9a activity, leading to decreased H3K9
epigenetic modifications of histone are reversible and highly gov- methylation levels. The increased acetylation level for residues
erned by three categories of epigenetic modulators: “writers”, proximal to H3K4 contributes to repressing LSD1 activity and
“erasers”, and “readers”. “Writers” mediate epigenetic modifica- subsequent H3K4 methylation enhancement, whereas sumoylated
tions transfer to the histone tails, while “erasers” remove such H4 stimulates intranucleosomal demethylation by LSD1 [11].
chemical modifications, and “readers” recognize and bind to these Given that various histone epigenetic modulators undergo
specific epigenetic marks. Histone acetyltransferases (e.g., CBP/ intimate interplay and act in a combined manner to activate or
p300) and histone methyltransferases (e.g., EZH2, G9a, and DOT1L) repress chromatin-mediated transcription, dual inhibitors target-
represent well-known “writers”. Histone deacetylases (e.g., HDAC, ing different epigenetic modulators is an effective strategy to
SIRT) and lysine demethylases (e.g., LSD1, JmjC KDM) represent improve single epigenetic-target agents' safety and efficacy and
“erasers”; “readers” largely fall into three families: bromodomain- overcome resistance [12,13]. In this review, we discussed the
containing proteins, methyl-lysine, and methyl-arginine-binding crosstalk of histone epigenetic modulators in regulating gene
domain-containing proteins, and PDH-containing proteins [2,3]. transcription, tumorigenesis and the recent development of dual
Epigenetic regulation plays a crucial role in many cellular pro- inhibitors targeting this crosstalk, compounds targeting multiple
cesses, including proliferation, differentiation, cell cycle, apoptosis, isoforms from the same family will not be discussed in this article.
and others. Dysfunction of epigenetic modulators is a common
feature of cancer development and progression, making these 2. Dual inhibitors targeting histone erasers and readers
epigenetic modulators attractive targets for cancer therapy [4e6].
Alternative to the irreversible genomic mutations that inactivate 2.1. Dual inhibitors targeting HDAC and BRD4
tumor suppressor genes or activate oncogenes in cancer, the
reversible nature of these epigenetic modifications confers a 2.1.1. Crosstalk between HDAC and BRD4
unique therapeutic advantage to targetingepigenetic modulators Histone deacetylases (HDAC) are a family of epigenetic enzymes
for cancer treatment. Aberrant histone modifications, including responsible that remove acetyl marks of lysine residues in histones
(de) acetylation and (de) methylation of lysine residues, are the and some non-histone proteins, including tumor suppressor p53,
most characterized and pharmaceutically used epigenetic changes. microtubules, and Hsp90 [14,15]. Histone lysine deacetylation re-
To date, six drugs targeting histone epigenetic modulators have stores lysine's positive charge and enhances histone's affinity for
been approved: five for HDAC inhibitors and one for EZH2 in- DNA, leading to a more condensed form of chromatin and gene
hibitors [7]. Furthermore, numerous small inhibitors are directed at silencing [16]. HDAC plays a crucial role in regulating multiple
distinct histone epigenetic modulators in clinical trials against processes of life, from cell cycle, cell proliferation, cell differentia-
various forms of cancer. A list of histone epigenetic modulators tion, and apoptosis to protein activities. Numerous correlative
under investigation is presented in Table 1. Despite the impressive studies have demonstrated aberrant HDAC expression in human
advances in epigenetic drug discovery, there are challenges and cancers, making HDAC an intently pursued target [17]. Starting with
limitations to overcome, including the following: (1) most Vorinostat (SAHA), the first HDAC inhibitor approved by the FDA
currently approved epigenetic inhibitors are restricted to treating (U.S. Food and Drug Administration) in 2006 for treating refractory
hematological malignancies, their efficacy in solid tumors remains cutaneous T-cell lymphoma, developing HDAC inhibitors for cancer
uncertain; (2) epigenetic agents often have a narrow safety margin, therapies has attracted tremendous efforts and resulted in
and dose-limiting adverse events such as the thrombocytopenia impressive successes with five HDAC inhibitors approved for clin-
and gastrointestinal toxicities are observed; (3) epigenetic agents ical use, four approved by FDA: Vorinostat (SAHA), Romidepsin
frequently require a longer time to exert their clinical response (FK228), Belinostat (PXD101), Panobinostat (LBH589), and one
compared with many other targeted agents; (4) intrinsic drug approved by CFDA (China Food and Drug Administration): Tucidi-
resistance to some epigenetic inhibitors such as HDAC and BRD4 nostat (also known as Chidamide). Also, other numerous HDAC
inhibitors have been described [8e10]. inhibitors are in the clinical trial stage [18].
Histone epigenetic modulators share many common residues; The canonical pharmacophore features of HDAC inhibitors
2
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Table 1
Approved and clinical trial drugs targeting histone epigenetic modulators.

Group Modulators Category Epigenetic target Approved Drugs Drugs in clinical trials
function

Histone HDACs Eraser Remove acetyl groups Tucidinostat (Chidamide), Abexinostat, Fimepinostat, Resminostat, Mocetinostat, Pracinastat,
Acetylation from histone lysines Vorinostat, Belinostat, Entinostat, Quisinostat, Givinostat, Tefinostat, Tinostamustine,
Romidepsin, Panobinostat Citarinostat, Ricolinostat, Domatinostat (4SC-202)
Trichostatin A, Bisthianostat, CUDC-101, OBP-801, CG200745, CXD101,
KA2507, OKI-179, NBM-BMX, AR-42,
SIRT1 Eraser Remove acetyl groups e Selisistat
from histone lysines
BET Reader Read acetyl groups on e BMS-986158, CPI-0610, GSK2820151, Mivebresib, MK-8628, Molibresib,
histone lysines Alobresib, RG6146, RVX000222, ZEN003694, ABBV-744
BRPF1 Reader Read acetyl groups on e e
histone lysines
CPB/p300 Writer Acetylate histone e CCS1477
lysines
Histone LSD1 Eraser Remove methylation e GSK2879552, IMG-7289, INCB0598725, TAK-418, ORY-2001, CC-90011,
marks from H3K4 and Seclidemstat (SP-2577),
Methylation H3K9
EZH2 Writer Methylates histone Tazemetostat CPI-0209, CPI-1205, GSK2816126, DS-3201b, HH2853, SHR2554, PF-
H3K27 06821497
EED Writer Methylates histone e MAK683
H3K27
DOT1L Writer Methylates histone e Pinometostat
H3K79
G9a Writer Methylates histone e e
H3K9
PRMT Writer Methylates histone e GSK3368715, GSK332659, JNJ-64619178, PF-06939999, PRT811
arginines

consist of three parts, as shown in Fig. 1: a surface recognition cap Other ZBGs, including 3-hydroxypyridin-2-thione [19] and dithio-
capable of interacting with the hydrophobic rim at the entrance of carbonate [20] are reported. Extensive structure-activity relation-
the active pocket, a zinc-binding group (ZBG) that chelates zinc ion ship (SAR) studies confirm that the cap group in HDAC inhibitors is
by forming coordinate bonds within the active site, and a linker that flexible and tolerates modification. Thus, ZBG can be easily linked to
connects the cap and ZBG and interacts with the hydrophobic other anticancer scaffolds providing numerous potent multi-target
tunnel of the active site. The most commonly used ZBG groups are inhibitors [12,21,22].
the hydroxamic acid and ortho-amino anilide group, while romi- The bromodomain and extra-terminal (BET) domain family are
depsin uses a thiol group to coordinate the active site's zinc ion. epigenetic readers featuring a tandem repeat of bromodomains

Fig. 1. Pharmacophore features and structures of approved HDAC inhibitors.

3
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

(BD1 and BD2) at their N-terminus. The BET family includes four stimuli, protein phosphatase PP1a dephosphorylated H3S10ph,
protein members (BRD2, BRD3, BRD4, and BRDT), and BRD4 is the which facilitated the class I HDAC-mediated deacetylation of
most characterized member of this family [23,24]. BRD4 prefer- nucleosomal H4K5ac/K8ac. This released chromatin-bound BRD4
entially binds acetylated lysine on histone's tail through their for subsequent recruitment of P-TEFb to promote the target gene
tandem bromodomains to recruit transcription factors, including P- expression [38]. In pancreatic ductal adenocarcinoma (PDAC) cell
TEFb, a global transcriptional elongation factor important for most lines, HDAC inhibitor 4SC-202 attenuated TGF b-induced gene
RNA polymerase II (Pol II) transcription. This (BRD4) also regulates repression and epithelial-to-mesenchymal transition in a BRD4-
the transcription of a vast array of inducible genes essential for and c-Myc-dependent manner [39].
multiple cellular processes [25,26]. BRD4 can also bind non-histone Recent studies revealed that the combination of HDAC and BRD4
proteins in a bromodomain-independent manner, such as the inhibitors showed synergistic anticancer activities in different tu-
binding of BRD4 to FLI1, MYB, SPI1, CEBPA/B, or p53 [27]. Moreover, mor cell models (Table 2). Fiskus et al. found that cotreatment with
BRD4 also shows an intrinsic kinase activity, a function that is not BRD4 antagonist, (þ)-JQ1, and HDAC inhibitor, Panobinostat, syn-
elucidated [28]. Deregulated BRD4 protein is strongly associated ergistically inhibited the growth of both cultured acute myeloid
with the occurrence and progression of several malignant human leukemia (AML) cells and primary AML blast progenitor cells
diseases, especially cancer. BRD4 is implicated in the expression of expressing mutant NPM1cþ or MLL fusion oncoprotein, but not of
many well-known oncogenes, including c-MYC, MYCN, FOSL1, and normal CD34cþ hematopoietic progenitor cells. Compared with
BCL-2, leading to considerable drug discovery efforts against this treatment with a single drug, combined treatment with (þ)-JQ1
target [29e31]. Currently, many BRD4 inhibitors have been and Panobinostat exerted a superior effect in prolonging the sur-
discovered and reported, and some of them are undergoing vival of non-obese diabetic/severe combined immunodeficient
different clinical trial phases for patients with solid tumors and (NOD/SCID) mice engrafted with OCI-AML3 or MOLM13 cells [40].
hematologic malignancies, such as BMS-986158, CPI-0610, Mive- Mazur et al. observed that combined inhibition of BRD4 and HDAC
bresib (ABBV-075), Alobresib (GS-5829), ZEN003694, ABBV-744, using (þ)-JQ1 and SAHA exerted a potent synergistic tumor-
and others [13,32,33]. Preliminary results from clinical trials suppressive effect on PDAC cells in vitro and in vivo, without evi-
confirm the anticancer potential of BRD4 inhibitors, but their effi- dence of tumor relapse or metastasis [41]. Geng's group showed
cacy as single agents against solid tumors seems to be limited. that HDAC inhibitors facilitated BRD4-mediated transcriptional
Frustratingly, intrinsic and acquired drug resistance and adverse upregulation of leukemia inhibitory factor receptor (LIFR) by
events have been observed [10,34,35]. increasing histone acetylation at the LIFR promoter. This led to
HDAC and BRD4 share many common acetylation marks of feedback activation of JAK1-STAT3 signaling and attenuated the
lysine residues on histone tails, such as H3K4 and H3K9. So, it is not efficacy of HDAC inhibitors in breast cancers. Conversely, blocking
surprising that there exists an intimate interplay between HDAC the LIFR-JAK1-STAT3 signal with BRD4 inhibitor potentiates HDAC
and BRD4. HDAC condenses chromatin structure by removing inhibitor activity in breast cancer [42]. Zhao's group also confirmed
acetylation “marks” on histone, which need to be recognized by the combining BRD4 and HDAC inhibition a potential therapy for breast
BRD4 before facilitating the activation of specific target genes. The cancer. Mechanistically, combination treatment with (þ)-JQ1 and
global acetylation profile of the genome regulated by HDAC is Mocetinostat attenuated the Ras/MEK/ERK pathway through up-
closely related to the recruitment and location of the BRD4 protein regulating USP17, leading to reduced breast cancer cell viability
(Fig. 2). [43]. Zhang et al. illustrated that Panobinostat and OXT-015 com-
Direct or indirect functional interplay between HDAC and BRD4 bination elicited a pronounced synergistic reduction in cellular
has been reported. Fu et al. demonstrated that N-CoR/HDAC3 viability across a broad range of glioblastoma (GBM) model systems
complex suppressed P-TEFb activity and consequently transcrip- and exerted antiglioma activity in GMB xenograft models [44].
tional elongation through interaction with HEXIM1 to deacetylate Meng et al. also found that cotreatment with Panobinostat and
cyclin-dependent kinase 9 (CDK9), an internal component of P- OXT-015 or (þ)-JQ1 caused improved repression of GBM-associated
TEFb, thus counteracting the BRD4-mediated transcriptional acti- oncogenic genes and induction of GBM-associated tumor-sup-
vation effect [36]. pressive genes compared with individual drug treatment, contrib-
Signal transducer and activator of transcription 5 (STAT5) are uting to a highly synergistic antiproliferative and proapoptotic
essential regulators of cell differentiation, proliferation, and sur- effect [45]. Zhao et al. demonstrated that co-administration of
vival. A research work from Rascle's lab indicated that BRD2 was BRD4 inhibitor with HDAC inhibitor synergized to reduce the
required to efficiently recruit the transcriptional machinery at expression of proliferative drivers such as c-Myc, cyclin D1, and
STAT5 to activate its target genes [37]. Following HDAC inhibitor NFkB, promote apoptosis and achieved superior anticancer effects
treatment, delocalization of BRD2 from soluble chromatin to against cutaneous T-cell lymphoma [46]. In B-cell lymphoma cells,
insoluble chromatin and subsequent STAT5-mediated transcription Romidepsin suppressed BCL6 function by promoting BCL6 acety-
suppression was observed. Chen's group identified that upon stress lation and subsequent BCL6 downregulation, resulting in

Fig. 2. Molecular crosstalk model between HDAC and BRD4. BRD4 recognizes acetylated lysine on the nucleosomal histones and recruits P-TEFb to facilitate RNA polymerase II-
dependent transcription elongation. HDAC can affect the location and functions of BRD4 by changing the global acetylation profile of nucleosomal histones.

4
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Table 2
Published combination trials of HDAC and BRD4 inhibitors for cancer treatment.

HDAC inhibitor BRD4 inhibitor Cancer type Reference

Panobinostat (þ)-JQ1 AML [40]


SAHA (þ)-JQ1 Murine lymphoma [52]
LBH589 I-BET151 Melanoma [51]
SAHA or Mocetinostat (þ)-JQ1 Breast cancer [42,43],
SAHA (þ)-JQ1 Pancreatic adenocarcinoma [41]
Romidepsin (þ)-JQ1 B-cell lymphoma [47]
JNJ-26481585 (þ)-JQ1 Rhabdomyosarcoma [48]
Romidepsin (þ)-JQ1 Urothelial carcinoma [50]
Panobinostat OTX015 or (þ)-JQ1 Glioblastoma [44,45],
SAHA (þ)-JQ1 Gallbladder cancer [49]
SAHA (þ)-JQ1 Cutaneous T-cell lymphoma [46]

significant cell apoptosis and cell cycle arrest. When used combined HDACs isoform-selectivity. Consistent with the observed in vitro
with (þ)-JQ1, strong synergistic effects on proliferation and effects, in human Capan-1 pancreatic cancer xenograft model,
apoptosis were observed [47]. The combined therapeutic efficacy of compound 1 exerted an excellent in vivo anticancer efficacy and a
(þ)-JQ1 with various HDAC inhibitors was also validated in rhab- tumor growth inhibition (TGI) of 69% (bid, 15 mg/kg) and 87.7% (bid,
domyosarcoma (RMS) cells [48]. An evident synergistic interaction 20 mg/kg), respectively, more potent than that of (þ)-JQ1
of various HDAC inhibitors (i.e., JNJ-26481585, SAHA, LBH589, and (TGI ¼ 41%) and SAHA (TGI ¼ 49%) used alone or in combination
MS275) and (þ)-JQ1 to trigger apoptosis in RMS cells was discov- (TGI ¼ 57%). This illustrates that simultaneously targeting HDAC/
ered. Notably, (þ)-JQ1 and JNJ-26481585 cooperatively suppressed BRD4 is beneficial over single-target inhibition. RT-qPCR, immune
colony formation and triggered cell death in an in vivo RMS model, fluorescence, and western blotting assays revealed that in tumor
more effective than either treatment alone. Liu et al. showed that tissues, c-Myc and CDC25b mRNA expression was downregulated.
simultaneous inhibition of BRD4 and HDAC with (þ)-JQ1 and SAHA In contrast, Ac-H3 and Ac-H4 expression was increased, indicating
synergistically suppressed gallbladder cancer cell lines' prolifera- that BRD4 and HDAC were effectively inhibited in vivo.
tion by downregulating BRD4 expression and blocking PI3K/AKT By rationally fusing the pharmacophore of (þ)-JQ1 with well-
and MAPK/ERK pathways. In in vivo efficacy evaluation of NOZ established HDAC inhibitors such as CI994, SAHA, and Panobino-
xenograft model, cotreatment with (þ)-JQ1 and SAHA exerted a stat, Fulda's group discovered several new HDAC/BRD4 dual in-
higher efficacy than (þ)-JQ1 or SAHA used alone [49]. Hoffmann's hibitors. Among them, compound 2 (termed TW09) obtained by
group identified a significant synergistic interaction between substituting the tert-butyl ester of (þ)-JQ1 by a class I selective
(þ)-JQ1 and Romidepsin to induce apoptosis in urothelial carci- HDAC inhibitor CI994, was identified as the most active compound
noma cell lines, with reduced toxic side effects in normal cells [50]. with IC50 values of 729 and 74 nM against BRD4 BD1 and BD2,
In addition, combined inhibition of BRD4 and HDAC also syner- respectively, displaying approximately 10-fold higher affinity for
gistically suppressed melanoma and c-Myc-induced murine lym- BRD4 BD2 than BD1. The binding constants determined by
phoma growth [51,52]. isothermal titration calorimetry showed that TW09 has a high
These findings suggest that the design of dual HDAC/BRD4 in- binding affinity to BRD4 BD1 (Kd ¼ 69 nM) similar to (þ)-JQ1
hibitors could be a potentially effective therapy for cancers, and (Kd ¼ 51 nM) but is less potent than (þ)-JQ1 in binding to BDR4 BD2
medicinal chemists are dedicating considerable efforts to develop (Kd ¼ 230 nM). TW09 was also a potent inhibitor of HDAC1 with an
dual HDAC/BRD4 inhibitors. IC50 value of 290 nM, more potent than the parental HDAC inhibitor
CI994, which had an IC50 of 960 nM under the same conditions
2.1.2. Reported HDAC/BRD4 dual inhibitors (Fig. 4) [54]. The co-crystal structure of TW09 with BRD4 BD1
As the first reported potent pan-BET inhibitor, (þ)-JQ1 has been revealed that TW09 bound to BRD4 BD1 in a very similar pattern to
widely used as a tool molecule or positive control to explore BET (þ)-JQ1. TW09 formed key hydrogen-bond interactions between
proteins' biological function [24]. The co-crystal structure of triazole moiety with evolutionarily conserved residue Asn140,
(þ)-JQ1-BRD4 complex showed that the tert-butoxycarbonyl group leaving the HDAC inhibitor pharmacophore in a solvent-exposed
was oriented toward the solvent region, making it suitable for region, possibly accounting for its potent HDAC/BRD4 dual inhibi-
chemical modification, which extensive SAR explorations have tion potency [55]. In RMS cells, TW09 could efficiently modulate
validated. Sheng's group discovered a series of novel HDAC/BRD4 the expression of BRD4 and HDAC target proteins, including c-Myc
dual inhibitors, where hydroxamic acid moiety as the ZBG was and Ac-H3, induce apoptosis, block colony formation, and display
introduced to the pharmacophore of (þ)-JQ1 by replacing the tert- similar antiproliferative activity at the same concentration
butoxycarbonyl with a N-phenylacetamide (Fig. 3) [53]. Among compared with (þ)-JQ1 and MS-275 used in a 1:1 combination.
them, compound 1 exhibited potent and well-balanced inhibitory Further antitumor mechanism studies showed that TW09 triggered
activities against BRD4 BD1 (IC50 ¼ 11 nM) and HDAC1 significant mitochondrial apoptosis by up-regulating proapoptotic
(IC50 ¼ 21 nM), which was translated into its greater anticancer proteins (BIM, NOXA, PUMA, and BMF), downregulating anti-
potency against capan-1 cells (IC50 ¼ 0.15 mM) than (þ)-JQ1 apoptotic protein BCL-XL, and then activating BAK, BAX, and cas-
(IC50 ¼ 8.6 mM) and SAHA (IC50 ¼ 5.4 mM) used alone or in com- pase3/9, thereby exerting its potent anticancer activity. TW09 also
bination (IC50 ¼ 1.0 mM) by synergistic HDAC1/BRD4 dual inhibi- displayed robust antiproliferative activity against a panel of
tion. Compound 1 displayed pan-BET inhibitory activities with IC50 pancreatic cancer cell lines (MIA PaCa-2, DAN-G, and HPAC), more
values in the nanomolar range and showed a high selectivity potent than (þ)-JQ1, CI994 used alone or incombined treatment
against four tested non-BET BRDs, similar to that of (þ)-JQ1. HDAC [55]. More significantly, compared with single-agent or combined
profiling revealed that compound 1 exhibited a robust inhibition treatment with (þ)-JQ1 and CI994, TW09 showed more sustained
against the other four tested HDAC isoforms (HDAC2, 3, 6, and 8) growth-inhibitory effects in a long-term proliferation assay. Inter-
with IC50 values ranging from 21 to 192 nM, showing no obvious estingly, TW09 shared a very similar chemical structure with

5
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 3. HDAC/BRD4 dual inhibitor based on (þ)-JQ1.

Fig. 4. HDAC/BRD4 dual inhibitor based on (þ)-JQ1 and CI994.

compound 1. Structurally, the only difference between these two (IC50 ¼ 0.45 mM), SW620 (IC50 ¼ 1.78 mM), and DLD1
molecules is replacing the hydroxamic acid group in compound 1 (IC50 ¼ 2.11 mM), better than SAHA and RVX208 used alone or
with an ortho-amino anilide moiety. However, TW09 demon- combined. Compound 4 efficiently regulated c-Myc and Ac-H3
strated class I HDAC isoform selectivity, while compound 1 was a expression, blocked the cell cycle, promoted apoptosis, and
potent pan-HDAC inhibitor. induced autophagy in HCT-116 cancer cells. In the HCT-116 xeno-
Recently, He's group designed and synthesized several selective graft model, oral administration of compound 4 effectively inhibi-
HDAC/BRD4 dual inhibitors based on thieno[2,3-d]pyrimidine ted HDAC/BRD4, induced autophagic cell death, suppressed IL6-
compound 3, a novel BRD4 inhibitory scaffold (IC50 ¼ 0.85 mM) JAK-STAT signaling pathways, and showed superior in vivo anti-
discovered by fragment-based design strategy (Fig. 5) [56]. The tumor potencies than SAHA with TGI of 42.7% (15 mg/kg) and 68.8%
most potent compound, 4, simultaneously inhibited BRD4 and (30 mg/kg), respectively. No significant signs of toxicity were
HDAC2/6 with IC50values of 710 nM, 58 nM, and 73 nM, respec- observed in the main organs. Also, pharmacokinetics and pre-
tively. SAR analysis indicated that the connection position of ZBG, liminary safety studies showed that compound 4 had a reasonable
not the ZBG and linker moiety, was crucial for BRD4 inhibitory pharmacokinetic profile and oral bioavailability of 40.5% in rats.
activity. For example, compound 5 with the ZBG moiety on the ABBV-744 is a highly potent BD2-selective BRD4 inhibitor with
piperidine ring was inactive against BRD4. HDAC isoform selectivity Kd values of 3300 nM and 2.1 nM for BRD4 BD1 and BD2, respec-
assay showed that compound 4 exhibited good selectivity toward tively, being tested in phase I trial for relapsed/refractory AML
class I and class IIb HDAC (IC50: 0.05e0.97 mM) over class IIa and IV cancer and myelofibrosis [57,58]. The co-crystal structure of ABBV-
HDAC (IC50 > 10 mM). Compound 4 also displayed a high selective 744 with BRD2 revealed that the ethyl amide moiety is located at
profile toward BRD4 over other isoforms of BRD proteins. At the the solvent-oriented region and plays an essential role in differ-
cellular level, compound 4 potently suppressed the growth of three entiating BD1 and BD2 together with 2,6-dimethylphenyl ether
tested colorectal carcinoma cell lines including HCT-116 moiety.

6
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 5. HDAC/BRD4 dual inhibitors based on thieno[2,3-d]pyrimidine BRD4 inhibitors.

Inspired by these pieces of evidence, Hu's group designed and BET295, have been described as BET inhibitors. In this compound,
synthesized various novel HDAC/BRD4 dual inhibitors by merging the THQ acetamide motif exactly mimics the acetyl-lysine substrate
the common pharmacophore of HDAC inhibitors to the ethyl amide in binding with BRD4. The 4-isopropylcarbamate substituent forms
moiety of ABBV-744 (Fig. 6) [59]. Interestingly, by introducing a hydrophobic interaction with the WPF shelf region, and the para-
different zinc-binding groups to ethyl amide moiety and exploring position of the 6-arylsubstituent, which protrudes into the ZA
substituents on the 2-position of the phenyl ring, three dual in- channel, is solvent-exposed. Thus, compound 9 was designed as a
hibitors with different subtype selectivity were identified. Among dual HDAC/BRD4 inhibitor by incorporating the polymethylene
them, compound 6 simultaneously inhibited HDAC1 hydroxamate chain typical of SAHA to the para-position of the 6-
(IC50 ¼ 59.7 nM), BRD4 BD1 (IC50 ¼ 5.6 nM), and BRD4 BD2 arylsubstituent (Fig. 7). Compound 9 showed potent inhibition
(IC50 ¼ 2.1 nM) but possessed no obvious activity against HDAC4/6/ against BRD4 (IC50 ¼ 50 nM) and HDAC1/6 (IC50 ¼ 250 nM and
8/11 (IC50 > 10 mM), thereby defined as a HDAC1/BRD4 dual in- 420 nM). In cells, compound 9 inhibited cell proliferation in NUT
hibitor. Consistent with its HDAC subtype selectivity, compound 6 midline carcinoma (NMC) and AML cells and reduced TNF-a and IL-
significantly upregulated the expression level of Ac-H3 in MV-4- 6 production from LPS-stimulated peripheral blood mononuclear
11 cells, rather than Ac-tubulin, a physiological substrate of HDAC6. cells.
Also, compound 6 could potently attenuate c-Myc expression in Dimethylisoxazole is another well-known effective scaffold of
MV-4-11 cells, leading to G0/G1 cell cycle arrest, apoptosis induc- BRD4 inhibitor [60]. As a good starting point, several novel 3,5-
tion, and stronger inhibitory activity (IC50 ¼ 40 nM) than MS-275 dimethylisoxazole derivatives with a hydroxamate group were
(IC50 ¼ 260 nM), ABBV-744 (IC50 ¼ 300 nM), and OTX-015 synthesized and identified as HDAC/BRD4 dual inhibitors by Chen's
(IC50 ¼ 150 nM). Compound 7 was identified as a pan-HDAC/ group [61]. Considering the similarity between the ZA channels of
BRD4 dual inhibitor, exhibiting nanomolar potency against BRD4 and tubular channel of HDAC, the hydroxamic acid group was
HDAC1/3/6/8, BRD4 BD1 (IC50 ¼ 29.5 nM), and BRD4 BD2 connected to the 1-position of BRD4 inhibitor compound 10
(IC50 ¼ 6.0 nM). In MV-4-11 cells, compound 7 showed comparable through a flexible carbon chain, hoping to increase the ability to
antiproliferative activity, apoptosis induction, and cell cycle- occupy the ZA channel while achieving HDAC inhibitory activity
blocking effect compared with compound 6. Compound 8 showed (Fig. 8). Two representative compounds, 11 and 12 showed robust
a good potency for HDAC6 (IC50 ¼ 17.2 nM) and BRD4 BD2 inhibitory activity against HDAC1 (IC50 ¼ 0.27 and 0.15 mM,
(IC50 ¼ 1.2 nM), with a 13- and 300-fold selectivity over HDAC1 respectively) and BRD4 (IC50 ¼ 0.85 and 0.67 mM, respectively).
(IC50 ¼ 228.3 nM) and BRD4 BD1 (IC50 ¼ 368.7 nM), respectively, Temperature shift experiments showed that compounds 11 and 12
making it a HDAC6/BRD4 BD2 dual inhibitor. Compound 8 had a good affinity with BRD4 and exhibited a certain selectivity for
demonstrated potent inhibitory activity against MV-4-11 cells with the other five tested bromodomain proteins. In vitro anticancer
an IC50 value of 0.3 mM; however, unlike compounds 6 and 7, it assays revealed that compounds 11 and 12 strongly repressed the
could not effectively induce G0/G1 phase arrest at a concentration growth of K562 and MV4-11 cancer cell lines, with IC50 values of
of 1.0 mM, similar to the BD2-selective BET inhibitor ABBV-744. 0.91e2.33 mM, slightly weaker than the positive control drugs,
Several tetrahydroquinoline (THQ) derivatives, including I- (þ)-JQ1 and SAHA.

7
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 6. HDAC/BRD4 dual inhibitors based on ABBV-744.

Fig. 7. HDAC/BRD4 dual inhibitors based on I-BET295.

Recently, Liu's group also reported a series of novel was the most active against BRD4 with 88% inhibition at 10 mM.
dimethylisoxazole-based HDAC/BRD4 dual inhibitors by merging Compound 14 also showed potent inhibition against HDAC1,
the core structural scaffold of a reported BRD4 inhibitor, P-0014, HDAC2, and HDAC3, rather than HDAC6 (IC50 > 1.0 mM), with IC50
and an HDAC inhibitor compound 13 (Fig. 8) [62]. Compound 14 values of 0.18 mM, 0.30 mM, and 0.005 mM, respectively. In THP-

8
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 8. HDAC/BRD4 dual inhibitors based on dimethylisoxazole BRD4 inhibitors.

1 cells, compound 14 dose-dependently downregulated c-Myc compound 16 showed a more dramatic down-regulation effect on
expression and promoted Ac-H3 expression, indicating that com- c-Myc level than SAHA and (þ)-JQ1 in OCI-AML2 and OCI-AML3
pound 14 was an effective HDAC/BRD4 dual inhibitor at the cellular cells at 1.0 mM [64].
level.
Based on the reported N6-benzoyladenine BRD4 inhibitors, 2.2. Dual inhibitors targeting HDAC and BRPF1
Seika et al. designed and synthesized various novel HDAC/BRD4
dual-target inhibitors (Fig. 9) [63]. Compound 15 showed the BRPF1 (the bromodomain- and PHD finger-containing protein 1)
strongest HDAC/BRD4 dual-inhibitory activity, with IC50 values of belongs to the subfamily IV of the human bromodomains and is a
0.26 and 2.7 mM. In HL-60 cancer cells, compound 15 can inhibit cell unique multivalent epigenetic “reader” harboring three histone-
proliferation (GC50 ¼ 4.4 mM), time-dependently induce apoptosis, binding domains: one bromodomain for recognizing acetyl-lysine
and enhance ATRA-induced cell differentiation (EC50 ¼ 9.4 mM). marks, two PHD fingers for binding to unmodified histone H3,
Importantly, compound 15 also exhibited growth-inhibitory activ- and a PWWP domain for specific interaction with methylated his-
ity against several BRD4 inhibitor-resistant cancer cells, including tone H3. BRPF1 associates with three histone acetyltransferases,
K-562 (GC50 ¼ 7.1 mM), T-47D (GC50 ¼ 2.3 mM), and OVCAR-5 including MOZ (monocytic leukemic zinc finger), MORF (MOZ-
(GC50 ¼ 18 mM) cell lines. related factor), and HBO1 (HAT bound to ORC1), assembling a
Shao et al. discovered various novel HDAC/BRD4 dual inhibitors, tetrameric acetyltransferase complexe that acetylate histones and
where the hydroxamic acid moiety as the zinc-binding functional regulate transcriptional programs. In the tetrameric complex,
group was introduced to the free hydroxyl group of the BRD4 in- BRPF1 serves as a central scaffolding protein to link subunits
hibitor, RVX-OH, through various linkers (Fig. 9). The most prom- together into the functional complex, restrict substrate specificity,
ising compound, 16, had an IC50 value of 0.20 mM against HDAC1. and enhance acetyltransferase activity [65]. Various mutations in
Compound 16 also showed potent activity toward BRD4 BD2 BRPF1 have been implicated in acute leukemias, and therapeutic
(IC50 ¼ 0.40 mM), but not BRD4 BD1 (IC50 > 5.0 mM). Compound 16 targeting of BRPF1 by small molecules is emerging as an attractive
showed efficient antiproliferative activities against three AML cell strategy for AML therapy [66].
lines (MV4-11: IC50 ¼ 0.56 mM, OCI-AML2: IC50 ¼ 0.38 mM, and OCI- Compounds 17e18 were synthesized and identified as potent
AML3: IC50 ¼ 0.43 mM) with greater potency than the reference BRPF1 inhibitors starting with the fragment hit N-methylquinolin-
drugs, RVX-208 and SAHA. Western blotting revealed that 2(1H)-one, which had an IC50 of 4.8 mM for BRPF1 [67]. Compounds
9
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 9. Other reported HDAC/BRD4 dual inhibitors.

17e18 potently inhibited BRPF1 with IC50 values of 7.9 and 60 nM, pharmacological inhibition of LSD1 demonstrated anti-
respectively. Co-crystal structure of 18 with BRPF1B displayed that proliferation and anti-metastasis effects in diverse cancers.
compound 18 occupied the conserved acetyl methyl pocket of Studies from Shi's group indicate that genetic or pharmacological
BRPF1 and the 1,4-dimethylquinolin-2-one scaffold served as the abrogation of LSD1 can elicit potent antitumor immunity and T-cell
acetyl-lysine mimetic with the carbonyl forming a hydrogen bond infiltration in low or non-immunogenic tumors. This is done
with the conserved Asn708 and forming an indirect hydrogen bond through an induced expression of endogenous retroviruses and
with Tyr665 mediated by a water molecule. Structure guided reduced expression of the RNA-induced silencing complex [84].
optimization of benzhydroxamic acids by Heimburg et al. led to a Therefore, developing small-molecule inhibitors of LSD1 has been
series of highly potent and selective HDAC8 inhibitors [68]. Com- an attractive area of anticancer drug discovery [78], and numerous
pounds 19 was one of the most potent and selective HDAC8 in- LSD1 inhibitors have been developed over the last decade.
hibitor with an IC50 value of 69 nM. Detailed SAR exploration Till date, seven LSD1 inhibitors, including IMG-7289,
showed that the two-atom linkers between benzhydroxamic acid GSK2879552 [76], TAK-418 [6], ORY-2001 [85], INCB059872
moiety and aromatic cap group and the meta-substitution pattern [86,87], CC-90011 [88], and SP-2577 (Seclidemsta) [89], are being
were crucial for the HDAC8 selectivity. tested in I/II phase trial for AML and solid tumors (Fig. 11) [90,91].
Based on the available co-crystal structure of BRPF1 and com- IMG-7289, GSK2879552, TAK-418, ORY-2001, and INCB059872 are
pound 18, and the reported selective HDAC8 inhibitors 19, com- irreversible LSD1 inhibitors derived from tranylcypromine (TCP or
pounds 20e21 were designed and synthesized using a PCPA), a mechanism-based covalent monoamine oxidase MAO in-
pharmacophore fusion approach (Fig. 10). The biochemical and hibitor with moderate activity against LSD1. Mechanistically, TCP-
biophysical assay identified compounds 20 and 21 as potent dual based LSD1 inhibitors inactivated LSD1 through covalent modifi-
HDAC/BRPF1 inhibitors with inhibitory activities against HDAC8 cation of FAD in the amine oxidase domain active pocket. CC-90011
(IC50 ¼ 443 and 956 nM, respectively) and BRPF1 (Kd ¼ 67 and and SP-2577 are highly potent reversible LSD1 inhibitors developed
4080 nM, respectively) [69]. Unfortunately, compounds 20 and 21 by Celgene Corporation and Salarius pharmaceuticals, respectively.
showed disappointing anticancer activity against two tested AML Recent structural and functional studies have uncovered the
cell lines, THP-1 and HL-60. This may be due to their poor cell coordinated crosstalk between LSD1 and HDACs. First, LSD1 and
membrane permeability. HDAC1/2 often function as a catalytic component in the same
transcriptional repressor complexes, including NuRD (nucleosome
3. Dual inhibitors targeting histone erasers remodeling and deacetylase), CoREST (corepressor of RE1 silencing
transcription factor) [92], and Sin3A (SIN3 transcription regulator
3.1. Dual inhibitors targeting LSD1 and HDAC family member A), and exert their demethylase and deacetylase
activities in a mutually dependent manner. Using real-time NMR
3.1.1. Crosstalk between LSD1 and HDAC assay, Song et al. confirmed that LSD1 tethered HDAC1 by RCOR1
LSD1 (lysine-specific demethylase 1), also known as KDM1A, is scaffold protein to form a stable ternary CoREST complex and the
the first discovered histone demethylase that requires FAD as a two enzymes were functionally related. However, they are located
cofactor to exert its demethylase activity. LSD1 specifically erases at opposite ends of the complex. Inhibiting or activating one
methylation marks from mono- and dimethylated lysine 4 in his- enzyme significantly influenced the activity of the other enzyme
tone 3 (H3K4me1/2), leading to transcriptional silencing [70]. By [93]. LSD1 regulated embryonic stem and carcinoma cells' plurip-
interacting with androgen receptor or estrogen receptors, LSD1 can otency through HDAC1 mediated deacetylation of H4K16 interde-
demethylate mono- and dimethylated lysine 9 in histone 3 pendently through CoREST complex. LSD1 coordinates with the
(H3K9me1/2), thereby promoting gene transcription. LSD1 also SIN3A/HDAC complex to repress a series of proapoptotic genes and
demethylate nonhistone substrates, such as p53 [71], DNMT1 (DNA maintain sensitivity to chemotherapy in breast cancer [94].
methyltransferase 1) [72], E2F1 (E2F transcription factor 1) [73], Second, the direct interaction of HDACs with LSD1 in the cell
and MYPT1 (myosin phosphatase targeting subunit 1) [74]. LSD1 is nucleus has been confirmed. HDAC1 deacetylated LSD1 at K374 in
significantly overexpressed or dysregulated in multiple types of the substrate-binding domain, subsequently facilitated the LSD1/
cancer, such as prostate cancer [75], small cell lung cancer [76], H3 effective binding and promoted its demethylation activity,
colon cancer [77], breast cancer [78], and AML [79,80]. It has been leading to a more permanent silencing of target genes (Fig. 12A)
implicated in many cellular processes such as EMT (epithelial- [95]. Huang et al. demonstrated that LSD1 and HDAC5 were coor-
mesenchymal transition) [81], cell proliferation and differentiation, dinated overexpression in human breast cancer cell lines and
control of stemness [82], autophagy [83], and malignant trans- clinical patient samples. HDAC5 physically interacted with LSD1
formation. Also, high LSD1 levels frequently correlate with poor and promoted LSD1 stability and demethylation activity through
clinical outcomes in various cancers. RNAi-mediated knockdown or up-regulating USP28, a bona fide deubiquitinase of LSD1, thereby
10
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 10. Reported HDAC/BRPF1 dual inhibitors.

Fig. 11. LSD1 inhibitors undergoing clinical trials.

11
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

facilitating breast cancer progression (Fig. 12B). Conversely, siRNA LSD1 inhibitor, SP-2509, in inhibiting AML cell growth (both
depletion of HDAC5 significantly downregulated LSD1 protein cultured and primary AML blasts). Simultaneous HDAC and LSD1
stability and suppressed breast cancer cell proliferation and inva- inhibition with Panobinostat and SP-2509 showed greater efficacy
sion [96]. Inactivating HDAC5 with sulforaphane, a natural bioac- in prolonging the overall survival of mice bearing human AML xe-
tive HDAC inhibitor, also led to a profound, decreased expression of nografts compared with each drug alone, without any sign of
LSD1 protein through HDAC5 modulated LSD1 ubiquitination sys- toxicity [99]. A combination of GSK690 and JNJ-26481585 or SAHA
tem [97]. Additionally, the acetylation level for residues proximal to caused a synergistic induction of cell death in RMS cells by
H3K4 can differently influence LSD1 activity. The treatment of changing the balance between pro- and antiapoptotic BCL-2 pro-
cancer cells with HDAC inhibitors suppressed the expression of teins, thereby inducing the intrinsic mitochondrial apoptotic
LSD1 and promoted the H3K4 methylation and H3 acetylation. pathway [102]. Chandra's group conducted a preclinical evaluation
Third, HDAC inhibitors were capable of synergizing with LSD1 of combined HDAC and LSD1 inhibition in glioblastoma (GMB)
inhibitors to suppress various malignancies [98e100]. In MDA-MB- [103,104]. Results revealed that the simultaneous treatment of GBM
231 breast cancer cells, Huang and Co. found that shRNA-mediated cells with TCP and SAHA led to synergistic apoptotic cell death,
LSD1 depletion conferred enhanced antineoplastic effect of sulfo- while there were no significant effects on normal cells [104]. Also,
raphane. Cotreatment with sulforaphane and LSD1 inhibitor HCI- combining TCP and SAHA showed efficacy in decreasing the
2509 synergistically suppressed breast cancer cell line prolifera- viability of patient-derived glioma stem cells. Particularly, the
tion, including MCF10A-CA1a, MDA-MB-231, and MDA-MB-468, combination treatment could reduce tumor burden and extend
except for normal breast epithelial cells. Moreover, in a thymic overall survival in an orthotopic xenograft GBM mouse model. In
nude mice engrafted with MDA-MB-231 cells, combined therapy contrast, individual treatment with SAHA or TCP provided no
with sulforaphane and LSD1 inhibitor was significantly superior to improvement in overall survival [103]. Welch et al. identified a
treatment with sulforaphane or HCI-2509 alone in inhibiting tumor synergistic interaction of SP2509 with Romidepsin to inhibit the
growth [97]. Another research work from the same group showed growth of ewing sarcoma cell lines [100].
that combined treatment with LSD1 inhibitor, Pargyline, and HDAC
inhibitor, SAHA, also led to superior growth inhibition and 3.1.2. Reported LSD1/HDAC dual inhibitors
apoptotic death in triple-negative breast cancer cells [101]. Pan- Considering the intimate crosstalk between HDAC and LSD1 and
HDAC inhibitor, Panobinostat, interacted synergistically with the distinct benefits of combining HDAC inhibitors with LSD1

Fig. 12. Molecular crosstalk model between HDAC1/5 and LSD1. (A) HDAC1 deacetylates LSD1 at K374, which results in proper LSD1/H3 binding and demethylation of H3K4 to
repress target genes. HDAC inhibitors stimulate LSD1 acetylation, leading to decreased LSD1 activity, up-regulated H3K4 methylation, and derepression of target genes; (B) HDAC5
facilitates stability and demethylase activity of LSD1 through up-regulating the expression of LSD1 deubiquitinase USP28, while HDAC5 inhibitors promote LSD1 degradation
through down-regulating USP28 expression in an HDAC5-dependent manner.

12
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

inhibitors, developing inhibitors that simultaneously target HDAC expression of Corin-target genes was associated with prolonged
and LSD1 represents an interesting anticancer approach. To date, survival time in a DIPG patient cohort.
several dual LSD1/HDAC inhibitors have been reported by our and Our group reported various TCP derivatives as LSD1/HDAC dual
other groups, and most dual LSD1/HDAC inhibitors were designed inhibitors. Compound 23 emerged as the most promising dual in-
by tethering known hydroxamic acid or benzamide group of HDAC hibitor, with potent inhibitory activity against HDAC1/2 and LSD1
inhibitors to the covalent LSD1 inhibitor, TCP. with an IC50 value of 15 nM, 23 nM, and 1.20 mM, respectively
Corin (compound 22) is a potent LSD1/HDAC dual inhibitor (Fig. 14). Compound 23 could efficiently inhibit tumor growth in
obtained by fusing the pharmacophore of an HDAC inhibitor MGC-803, MCF-7, SW-620, and A-549 human cancer cell lines with
(Entinostat) and a TCP-based LSD1 inhibitor (Fig. 13) [105]. Corin IC50 values ranging from 0.81 to 4.28 mM by decreasing the mito-
simultaneously blocked the LSD1 and HDAC1 activity of the CoREST chondrial membrane potential and inducing cellular apoptosis. The
complex with an IC50 of 0.33 and 0.21 mM, respectively. Interest- intracellular LSD1 and HDAC inhibitory activity of compound 23
ingly, Corin displayed more robust and sustained inhibition of was proven by validating the significant accumulation of H3K4Me2,
HDAC1 than Entinostat alone or a combination of Entinostat and H3K9Me2, and Ac-H3 after treatment with compound 23 in MGC-
Bizine in the CoREST complex, but not in the MiDAC complex 803 cells [107].
lacking an LSD1 subunit. Though, Entinostat was a reversible HDAC A recent study by Sadhu et al. reported novel LSD1/HDAC6 dual
inhibitor, irreversible inhibition of HDAC1 by Corin was observed. inhibitors using TCP as an LSD1 inhibitor pharmacophore and
Corin exhibited more powerful antiproliferative activities against a hydroxamic acid as an HDAC inhibitor pharmacophore (Fig. 14)
panel of ten melanoma cell lines than MS-275 at 1.0 mM, especially [108]. Out of approximately 50 new compounds tested, compound
in WM983B MM cells (EC50 ¼ 95 nM), but showed no apparent 24 was identified as the most active dual inhibitor. Compound 24
effects on primary human melanocytes. Corin also possessed exhibited irreversible and potent LSD1-inhibitory activity
excellent antiproliferative potency against cutaneous squamous (IC50 ¼ 5 nM) and a high selectivity over the MAO-A
cell carcinoma lines IC1 (IC50 ¼ 41 nM) and MET1 (IC50 ¼ 6 nM), (IC50 ¼ 8600 nM) and MAO-B (IC50 ¼ 6600 nM). Compound 24
which was better than the mono-functional inhibitors or a com- also displayed nanomolar activity against HDAC6 and HDAC8 with
bination of MS-275 and LSD1 inhibitor, GSK2879552. Interestingly, an IC50 value of 48 and 71 nM, but had a low activity against other
depleting CoREST1 or LSD1 conferred enhanced anticancer effi- HDAC isoforms (1, 2, 3, 5, 7, 9, and 10), exhibiting 70-fold selectivity
ciency to MS-275 in WM983B and HCT-116 cells, but did not alter for HDAC6 against HDAC1 (IC50 ¼ 3400 nM). Pure enantiomers (1R,
Corin's antiproliferative effects. This suggests that Corin may also 2S and 1S, 2R) of compound 24 were also synthesized to assess the
exert its anticancer potency by blocking HDAC in other contents. difference in potency. Interestingly, they showed similar potency to
Corin displayed good metabolic stability in human plasma, human compound 24 in enzymatic potency and antiproliferative activity.
liver microsomes, and mouse liver microsomes. In SK-MEL-5 mouse Consistent with the authors' predictions, compound 24 showed
xenografts, treatment with Corin by ip daily administration at a remarkable antiproliferative activities against three multiple
dose of 30 mg/kg for 28 d, caused a 61% reduction in tumor volume myeloma cell lines, MM.1S (EC50 ¼ 2 nM), MM.1R (EC50 ¼ 6 nM),
without noticeable effect on body weight and blood cell counts, and RPMI-8226 (EC50 ¼ 280 nM). Pharmacokinetic studies in mice
while MS-275 was too toxic to experiment with the same dose. identified that compound 24 exerted good oral bioavailability of
Western blotting and qRT-PCR analysis of the tumor cells collected 44%. More significantly, compound 24 showed potent in vivo anti-
at the end of the animal studies showed that the expression of Ac- tumor efficacy in MM.1S xenograft models. Oral administration of
H3K9, H3K4me2, p21, CHOP, and MXD1 elevated in the Corin- compound 24 at 25 mg/kg for 12 d showed a good TGI of 67.0%
treated group. Also, immunohistochemistry staining revealed that without significant changes in body weight, which was superior to
the expression levels of Ki-67 were downregulated in the tumor single LSD1 inhibitor GSK-2879552 (25 mg/kg, PO, TGI ¼ 40%) and
tissues of the Corin treatment group. HDAC6 inhibitor ACY-1215 (50 mg/kg, IP, TGI ¼ 36%).
Furthermore, the significant antitumor efficacy of Corin was By incorporating SAHA and TCP with various polyamine chains,
also confirmed in HSJD-DIPG007 diffuse intrinsic pontine gliomas Milelli et al. synthesized various TCP-based LSD1/HDAC dual in-
(DIPG) xenografts by Shi and Co [106]. Co-targeting LSD1 and hibitors (Fig. 14). Compound 25 showed good dual-inhibitory ac-
HDACs with Corin synergistically changed global histone methyl- tivities with a Ki of 42.52 nM for HDAC1 and an IC50 value of
ation and acetylation and potently suppressed DIPG growth both 3.85 mM for LSD1. Compound 25 showed comparable cytotoxic
in vitro and in vivo by re-programming chromatin to inhibit cell activity against MCF-7 (IC50 ¼ 39.6 mM) compared with SAHA
cycle, while promoting death and differentiation. Impressively, the (IC50 ¼ 38.2 mM), while at 70.0 mM compound 25 displayed more

Fig. 13. Structure of LSD1/HDAC dual inhibitor, Corin.

13
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 14. Reported TCP-based LSD1/HDAC dual inhibitors.

pronounced anticancer effect than SAHA (68.6% vs 56.6% of dead possessed significant HDAC inhibitory activity against HDAC1,
cells) [109]. HDAC2, and HDAC6 with IC50 values of 0.37, 0.40, and 0.015 mM,
4SC-202 (compound 26, also named domatinostat) is an orally respectively. Notably, it showed over 20-fold selectivity against
available non-TCP-based LSD1/HDAC dual inhibitor and is currently HDAC6 over HDAC1/2. Compound 28 displayed potent in vitro
undergoing phase I clinical trials (Fig. 15). 4SC-202 exhibited se- anticancer potency against all tested seven gastric cancer cell lines
lective inhibitory activities on class I-selective HDAC with an IC50 with IC50 values of 0.23e1.56 mM, more potent than ORY-1001 and
value of 1.20 mM, 1.12 mM, and 0.57 mM for HDAC1, HDAC2, and SP-2509. In addition, compound 28 significantly modulated the
HDAC3, respectively [110]. It also displayed inhibitory activity expression of Bcl-2, Bax, Vimentin, ZO-1 and E-cadherin, induced
against LSD1 with an IC50 value of 1.20 mM [111]. Numerous studies apoptosis, reduced colony formation and suppressed migration in
have demonstrated that 4SC-202 exerted potent antitumor efficacy MGC-803 cancer cells. Preliminary ADME studies revealed that
against multiple cancer cell lines, including hepatocellular carci- compound 28 showed acceptable metabolic stability in human
noma cell [112], medulloblastoma cell [111,113], colorectal cancer liver microsomes with minimal inhibition of cytochrome P450s
cell [114], cutaneous T-cell lymphoma [115], oral squamous cell [118].
carcinoma [22], and urothelial carcinoma cell [110]. Moreover, in
both syngeneic mouse models with CTL-high C38 or CTL-low CT26 3.2. Dual inhibitors targeting LSD1 and JmjC KDM
tumors, treatment with 4SC-202 led to elevated expression of
cancer-germline antigens, antigen-presenting machinery genes, 3.2.1. Crosstalk between LSD1 and JmjC KDM
and MHC class I and II molecules, along with cytotoxic T-cells (CTL) The JmjC domain-containing histone lysine demethylases
infiltration, which reinforce immune responses against tumors. represent the largest lysine demethylase superfamily, consisting of
4SC-202 significantly reduced tumor volume in both CT26 and C38 20 members divided into five subfamilies (KDM2/7, KDM3, KDM4,
tumor models, whereas anti-PD-(L)1 antibody had no obvious tu- KDM5, and KDM6) [119]. Unlike LSD1, a FAD-dependent deme-
mor inhibitory effect. When anti-PD-(L)1 antibody was added, 4SC- thylase, JmjC KDM, catalyze oxidative demethylation reactions with
202 showed enhanced anticancer effects compared with single- Fe (II) and 2-oxoglutarate (2-OG) as cofactors. Mechanistically,
agent therapy and substantially prolonged event-free survival Fe(II)/2-OG complex reacts with molecular oxygen to yield carbon
[116]. Notably, all event-free animals were completely tumor-free dioxide, succinate, and reactive ferryl-oxo species, which hydrox-
at the end of the study. Currently, 4SC-202 and anti-PD-1 anti- ylates the methylated histone lysine residue to produce a hemi-
body Avelumab combination therapy, for patients with advanced aminal intermediate. The unstable hemiaminal products then
Merkel cell carcinoma progressing on anti-PD-(L) 1 is under phase spontaneously break into formaldehyde and the demethylated
II clinical trial. lysine, while regenerating the active Fe(II) center. In contrast to the
More recently, Our group designed and synthesized a series of restricted catalytic activity of LSD1 at only mono- and dimethylated
novel non-TCP based LSD1/HDAC bifunctional inhibitors by lysines, JmjC KDM can erase all three methyl marks on lysine res-
replacing the amidoxime group of stilbene-based LSD1 inhibitor 27 idues [120]. JmjC KDM are overexpressed in multiple malignancies,
reported by our group with a hydroxamic acid moiety [117]. The stimulate cancer cell proliferation and invasion, and facilitate effi-
representative compound 28 exhibited potent activity and high cient tumor growth, underscoring the potential of JmjC KDM as an
selectivity against LSD1 over MAOA/B with IC50 of 1.64 mM. It also anticancer target [121,122]. Recently, significant progress has been
14
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 15. Reported non-TCP-based LSD1/HDAC dual inhibitors.

made in developing JmjC KDM inhibitors; most of them are physically interacted with LSD1 within the NuRD complex and
mechanism-based 2-OG competitive inhibitors and Fe2þ chelators cooperatively regulated H3K4 demethylation. In the term of
[123]. However, although some representative JmjC KDM inhibitors mechanisms, KDM5B/LSD1/NuRD complex bound to target gene
showed potent anticancer activity in vivo, only a few of them have promoters via H3K4me3 marks, which was initially recognized by
been approved in preclinical or clinical trials due to poor selectivity KDM5B, and the methyl groups were then sequentially erased by
and toxicity. the joint demethylase activities of KDM5B and LSD1, leading to
JmjC KDM demethylates di- and tri-methylated marks on lysine, gene silencing [129]. Zhang et al. found that the co-overexpression
including H3K4 and H3K9, while LSD1 is active on mono- or di- of LSD1 and KDM6B was tightly related to a bad prognosis in pa-
methylated H3K4 and H3K9. Given the prevalence of H3K4Me3 tients with head and neck squamous cell carcinoma (HNSCC).
and H3K9Me3, there should exist a functional connection between Depleting LSD1 with siRNA potentiates KDM6B inhibitor activity in
LSD1 and JmjC KDM to accomplish H3K4 and H3K9 demethylation, HNSCC and vice versa. Combinational treatment with TCP and
and research findings have confirmed the cooperation between KDM6B inhibitor GSK-J1 synergistically impaired tumor growth
LSD1 and JmjC KDM to exert their demethylases activity. and progression in the 4NQO-induced HNSCC model and FaDu
In normal prostate and human prostate cancers, KDM4C co- xenograft model in nude mice by inhibiting cell proliferation and
localized and interacted with LSD1 and androgen receptor (AR) to inducing cell apoptosis. In the xenograft model, the group treated
assemble into a multiple-specificity demethylase complex, which with TCP plus GSK-J1 showed a higher progression-free survival
removed the repressive methyl marks from mono-, di- and trime- than the single-agent treatment group [130].
thylated H3K9 to facilitate AR-dependent gene transcription [124]. Altogether, given the recent findings of their functional rela-
KDM4C inhibitors were inactive against prostate LNCaP and colon tionship, co-inhibiting JmjC KDM and LSD1 as a novel potential
HCT116 cancer cells, but when combined with LSD1 inhibitor NCL- therapeutic strategy for cancers have drawn increased attention.
2, they synergistically inhibited cancer cell growth [125]. KDM4C
and LSD1 were co-overexpressed and exerted an overlapping 3.2.2. Reported LSD1/JmjC KDM dual inhibitors
oncogenic activity in human melanoma. KDM4C and LSD1 coop- Mai et al. have disclosed various dual LSD1/JmjC KDM inhibitors
eratively blocked oncogenic Ras- or Braf-evoked senescence by (pan-KDM inhibitors as called by the authors), which were
promoting the expression of E2F target genes, thus facilitating the designed based on the LSD1 inhibitor, TCP, and linked with the 2,2-
formation of aggressive melanomas. Pharmacological inhibition of bipyridine (29) or 5-carboxy-8-hydroxyquinoline (30) scaffolds,
LSD1 or KDM4C effectively suppressed tumor growth in patient- two known 2-OG competitive templates developed for JmjC KDM
derived xenograft model of Braf-V600-mutant melanomas by inhibition (Fig. 16) [131]. In all target compounds synthesized,
restoring senescence [126]. KDM4A and LSD1 were also identified compounds 32 and 33 showed the most potent LSD1 inhibitory
as AR coregulators to mediate AR-dependent transcription via activity with IC50 values below 1.0 mM and displayed greater than
histone lysine-demethylation activity in human bladder cancer 40-fold selectivity over MAO-B, but these compounds had unsat-
[127]. Recent studies from Ray et al. showed that KDM4B formed a isfactory activities against JmjC KMD with an exception for KDM4C
protein complex with AP-2 family transcription factor, TFAP2C and (IC50 ¼ 2.7 mM for 32 and IC50 ¼ 1.2 mM for 33). Compound 31,
LSD1, which was enriched at the promoters of crucial trophoblast having an IC50 value of 2.2 mM against LSD1, exerted low or sub-
genes to regulate active gene expression in trophoblast stem cells micromolar activity against all tested JmjC KDM, but showed little
[128]. Shang's group demonstrated that in breast cancers, KDM5B or no effect against other tested 2-OG enzymes lacking KDM
15
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

activity, such as hypoxia-inducible factor hydroxylases FIH and specifically catalyzes H3K9 to form H3K9me1 and H3K9me2, which
PHD2. Compounds 32 and 33 induced more remarkable apoptosis acts as a preferred binding site for heterochromatin protein 1 (HP1)
than single 29, 30, or TCP, or the combination of 29 and TCP in both that recruits transcriptional repressors to exert transcriptional
LNCaP prostate and HCT-116 colon cancer cells, with a concomitant silencing [134]. Apart from methylating histones, G9a has also been
increase in H3K4Me2 and H3K9Me2/3, indicating effective dual- described to methylate several non-histone proteins [135,136]. For
target engagement. Nevertheless, compounds 32 and 33 have lit- example, G9a can dimethylate the lysine-373 residue of tumor
tle or no apoptotic induction ability in mesenchymal progenitor suppressor gene p53, leading to transcriptional inactivation of p53
noncancerous cells, suggesting anticancer selectivity. [137]. Under hypoxia, G9a can also methylate reptin at lysine 67,
Research by Benedetti et al. showed that compound 33 regu- facilitating the recruitment of HDAC1 to hypoxia-responsive gene
lated ERa signaling in breast cancer by inhibiting LSD1 and KDM6A. promoters to suppress the transcriptional activity of HIF-1a [138].
In MCF-7 and tamoxifen-insensitive-ERa-positive BT474 cancer G9a plays an essential role in many biological processes, including
cells, compound 33 strongly downregulated ERa expression at embryonic development [139], DNA damage repair [140], cell
protein and mRNA levels, accompanied by a significant growth growth, and autophagy [141].
arrest and apoptosis. Conversely, compound 33 failed to reduce the Studies have reported that the expression levels of G9a are often
growth of triple-negative MDA-MB-231 cells, underscoring the upregulated in many human cancers [142], including colorectal
vital role of ERa regulation for its anticancer effect. Also, compared cancer [143], breast cancer [144], ovarian cancer [145], liver cancer
with SAHA, 33 displayed less cytotoxicity to normal HaCaT cells. Its [146], gastric cancer [147], hematological malignancies, and over-
in vivo efficacy was confirmed in both chicken embryo and mouse expressed G9a is associated with both the occurrence and survival
models and ex vivo in human breast cancer specimens. In the of various cancers. Moreover, high G9a expression levels positively
chicken embryo model, 33 reduced tumor size and abolished the correlate with poor survival [144,148e151]. Knockdown of G9a or
migratory potential of MCF-7 cells compared to tamoxifen. In hu- pharmacological inhibition of its activity significantly inhibited cell
man breast cancer specimens, compound 33 impaired cell growth proliferation and tumorigenesis [143,152]. Collectively, the crucial
and suppressed ERa expression, recapitulating the observed effects roles of G9a in cancers suggest that G9a is a potential therapeutic
in MCF-7 cells. Its low cytotoxicity to healthy cells from isolated target for cancers [153].
neighboring tissue relative to breast specimens was also observed. To date, several G9a inhibitors have been discovered and re-
Oral administration of 33 also displayed good in vivo anticancer ported [152,154]. The best-studied G9a inhibitors are derived from
activity with a TGI of 60% in MCF-7 xenografted nude mice without the first identified small molecule G9 inhibitor BIX01294 [155],
obvious toxicity [132]. which is featured a quinazoline-amine scaffold (Fig. 17). Extensive
SAR exploration of BIX01294 displayed that modifications on the C2
4. Dual inhibitors targeting histone erasers and writers position of the quinazoline ring were well tolerated. Also, G9a in-
hibitors with complete distinct skeleton have been identified, such
4.1. Dual inhibitors targeting HDAC and G9a as aminoindole A-366, 2,4-diaminopyrimidine EPZ035544, 2-
aminobenzimidazole BRD4770, and quinolone CM-272.
4.1.1. Crosstalk between HDAC and G9a Interestingly, G9a is shown to functionally interact with HDAC to
G9a, also known as euchromatic histone methyltransferase 2 coordinate histone methylation and acetylation, promoting tran-
(EHMT2) or lysine methyltransferase 1C (KMT1C), is the second scriptional silencing of target genes. G9a and HDAC share several
identified histone methyltransferase [133]. Using S-adenosylme- common substrates, including H3K9 and p53K373. Acetylation of
thionine (SAM) molecule as the donor of the methyl group, G9a H3K9 (H3K9Ac) is related to gene activation, while H3K9 me2/3

Fig. 16. Reported dual LSD1/JmjC KDM inhibitors.

16
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 17. Structures of representative G9a inhibitors.

represses gene expression. H3K9 in the acetylated state cannot be much lower cytotoxicity in HEK293 with an IC50 value of 19.95 mM,
further methylated by G9a, thereby contributing to indirect inhi- which is about ten times higher than that of SAHA (IC50 ¼ 2.48 mM)
bition of G9a. HDAC1 and G9a have been found in the same protein and BIX-01294 (IC50 ¼ 2.05 mM).
complexes, including CoREST, C-terminal binding protein (CtBP)
[156,157]. The positive regulatory domain I-binding factor 1 (PRDI-
4.2. Dual inhibitors targeting HDAC and EZH2
BF1) binds to the IFNB1 promoter and silences transcription in vivo
by recruiting G9a and HDAC2, and the presence of HDAC2 is
4.2.1. Crosstalk between HDAC and EZH2
necessary for G9a-mediated methylation [158]. The growth factor
EZH2 (zeste homolog 2 enhancer) is an essential histone
independent 1 (Gfi1) recruits G9a and HDAC1 to form a stable
methyltransferase and a catalytic subunit of PRC2 (polycomb
repressive complex, which binds to the promoter of p21Cip/WAF1,
repressive complex 2) [165]. EZH2 and two PRC2 core components,
leading to decreased expression of p21Cip/WAF1 in HL-60 cancer cells
EED (embryonic ectoderm development) and SUZ12 (suppressor of
[159]. Hu et al. reported that G9a and HDAC interacted with Snail2
zeste 12 protein homolog), primarily catalyze the trimethylation of
to form a multiprotein complex to exert the repression of E-cad-
histone H3 lysine 27 (H3K27 me3) to silence PRC2 target genes
herin transcription in hepatocellular [160] and lung carcinoma cells
involved in-cell proliferation, cell-cycle regulation, cell differenti-
[161], thereby promoting their migratory and invasive capacity.
ation, and self-renewal. Apart from its transcriptional repressor
Inactivating G9a or HDAC with small inhibitors could significantly
activity on chromatin, EZH2 also methylates non-histone targets to
prolong survival time in mice harboring Snail2-overexpressing
regulate downstream genes. For example, EZH2/PRC2 can meth-
cancer cells [160]. In breast cancer, Wang et al. found that HDAC1
ylate cardiac transcription factor GATA4 at Lys299, thereby
served as a co-repressor with G9a and Yin Yang 1 (YY1) to silence
decreasing p300-mediated GATA4 acetylation and attenuating
ferroxidase hephaestin, contributing to enhanced cellular labile
GATA4 transcription [166]. Overexpression and gain-of-function
iron and increased proliferation of breast cancer in vitro and in vivo
mutations of EZH2 are frequently documented in various solid
[144]. Research by Seo's group documented that G9a cooperated
and hematologic malignancies [167,168]. Also, elevated EZH2
with both HDAC1 and HDAC2 and YY1 to repress JAK2 expression
transcript and protein levels are highly associated with tumori-
during ATRA-mediated leukemia cell differentiation [162]. Also,
genesis, progression, and poor prognosis, making it a promising
HDAC1 is functionally involved in G9a-mediated UHRF1 tran-
epigenetic target for cancer therapy [169,170].
scriptional repression in 12-O-tetradecanoylphorbol-13-acetate
Currently, numerous small-molecule EZH2 inhibitors have been
(TPA)-treated leukemia cells [163].
developed [171]. Tazemetostat (EPZ-6438) was approved in January
2020 by the FDA for metastatic or locally advanced epithelioid
4.1.2. Reported HDAC/G9a dual inhibitors sarcoma [170]. GSK2816126, CPI-1205, CPI-0209, SHR2554,
Given the intimate association between G9a and HDAC in HH2853, DS-3201b, and PF-06821497 are undergoing phase I/II
regulating tumor suppressor genes, simultaneously targeting G9a clinical trials as monotherapy and combined treatment for several
and HDAC may improve cancer therapy. Inspired by these pieces of types of cancer (Table 3). Interestingly, most of the best charac-
evidence, various novel G9a/HDAC dual inhibitors were designed terized EZH2 inhibitors are pyridone-based compounds, typically
and synthesized by integrating the hydroxamic acid group into the featured with a pyridone main pharmacophore as the “head” re-
C2 position of BIX01294 (Fig. 18) [164]. Among those synthesized gion, an amide linker as the “neck” region, different aromatics as
target compounds, 34 was the most active compound against G9a the “body” region, and variable “tail” structures linked to the bot-
with an IC50 value of 7.14 mM determined using in-cell Western tom of the body. The pyridone moiety provides essential in-
blotting. Compound 34 also showed significant HDAC inhibition teractions with the EZH2 domain in a SAM-competitive manner.
with IC50 values of 13.8 and 5.73 mM against HeLa nuclear extracts The“tail” structure was located in the solvent-exposed region,
and K562 nuclear extracts, respectively. Compound 34 effectively making it a suitable position for further derivatization (Fig. 20)
inhibited the proliferation of MDA-MB-231, MCF-7, and A549 with [172].
IC50 values ranging from 10.02 to 37.36 mM, which is less potent Accumulating pieces of evidence showed that EZH2 cooperated
than SAHA and BIX-01294. Notably, compound 34 also displayed with HDAC to reprogram histone methylation and acetylation,
17
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 18. HDAC/G9a dual inhibitor based on BIX02194.

Table 3
Small-molecule EZH2 inhibitors undergoing clinical trials.

Drugs Structure Phase Disease Trial Number Status

Tazemetostat (EPZ-6438) Approved Advanced or metastatic epithelioid sarcoma NCT04204941 Completed

GSK2816126 (GSK126) Phase I Elapsed/refractory DLBCL NCT02082977 Terminated


Transformed follicular lymphoma
Non-Hodgkin's lymphomas
Multiple myeloma

CPI-1205 Phase I B-cell lymphomas NCT02395601 Completed


Phase I Advanced Solid tumors NCT03525795 Completed
Phase Ib/II Metastatic castration resistant prostate cancer NCT03480646 Active, not recruiting

CPI-0209 Structure not in public domain Phase I/II Advanced solid tumors NCT04104776 Recruiting
PF-06821497 Phase I Relapsed/Refractory small cell lung cancer NCT03460977 Recruiting
Castration resistant prostate cancer
Follicular lymphoma

Valemetostat (DS-3201b) Phase I Acute myelogenous leukemia or Acute lymphocytic leukemia NCT03110354 Recruiting
Phase I/II Recurrent small cell lung cancer NCT03879798 Recruiting
Phase II Relapsed or refractory adult T-cell Leukemia/Lymphoma NCT04102150 Active, not recruiting
Phase I Lymphomas NCT02732275 Recruiting
Phase II Relapsed/Refractory peripheral T-cell lymphoma NCT04703192 Not yet recruiting
Phase IIb Metastatic prostate, urothelial and renal cell cancers NCT04388852 Recruiting
SHR2554 Structure not in public domain Phase I/II Metastatic castration resistant prostate cancer NCT03741712 Recruiting
Phase I/II Advanced solid tumors NCT04407741 Recruiting
B-cell lymphomas
Phase I Relapsed or refractory mature lymphoid neoplasms NCT03603951 Unknown
HH2853 Structure not in public domain Phase I Non-hodgkin's lymphomas NCT04390737 Recruiting
Advanced solid tumors

leading to more permanent silencing of target genes. EZH2 was tumors compared with the ARID1A wild-type ovarian tumors
shown to recruit HDAC1/2 to PRC2 through the EED protein, and (Fig. 19) [168]. Moreover, many synergistic antitumor effects have
interdependently mediated transcriptional repression [173]. HDAC been observed for EZH2 inhibitors combined with HDAC inhibitors.
inhibitors significantly attenuated EZH2 expression in both In EGFR-wild-type and EGFR-mutant NSCLC cells, cotreatment with
cultured and primary AML cells, leading to decreased levels of EZH2 inhibitor, 3-deazaneplanocin A, and HDAC inhibitor, SAHA,
H3K27Me3/2 and hyperacetylation of H4, with a loss of clonoge- synergistically reduced EZH2 and SUZ12 expression, strongly
netic survival [174]. Fukumoto et al. found that in ARID1A-mutant blocked the EGFR signaling pathway, and demonstrated superior
ovarian cancers, HDAC2 interacted with EZH2 to repress the in vivo antitumor efficacy than each inhibitor alone in EGFR-
expression of EZH2/ARID1A target proteins, including PIK3IP1, an tyrosine kinase-resistant H1975 xenograft model [175]. Pollock
inhibitor of PI3K-AKT signaling path. Also, pan-HDAC inhibitor, and Co. demonstrated similar EZH2 depletion upon HDAC inhibi-
SAHA, treatment conferred enhanced tumor growth suppression tion in epithelioid sarcoma [176]. Lue et al. documented co-
and improved survival to mice bearing ARID1A-mutant ovarian exposure to Romidepsin and diverse EZH2 inhibitors (GSK126,

18
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 19. Molecular crosstalk model between HDAC and EZH2 in ARID1A-mutant ovarian cancers. In ARID1A-mutated ovarian cancer cells, HDAC2 interacts with EZH2 to suppress
the expression of EZH2/ARID1A target tumor suppressor genes such as PIK3IP1, leading to enhanced cell proliferation and reduced apoptosis. HDAC2 inhibition or knockdown
derepresses the expression of PIK3IP1, inhibits cell growth and induces apoptosis.

EPZ011989, and CPI-1205) in lymphoma cell lines with EZH2 dys- [180,181]. Compound 36 showed EZH2 inhibitory activity compa-
regulation displayed potent synergistic anticancer activity. Mech- rable to compound 35, with an IC50 value of 7.37 mM. Concerning
anistically, Romidepsin-GSK126 cotreatment disrupted the HDAC inhibition, compound 36 displayed nanomolar potency
assembly of PRC2 complex by acetylating RbAp46/48, a subunit against HDAC6 (IC50 ¼ 5 nM) and sub-micromolar potency against
responsible for PRC2 complex recruitment to nucleosomes, leading HDAC1 (IC50 ¼ 0.43 mM), HDAC3 (IC50 ¼ 0.45 mM), and HDAC8
to decreased trimethylation of H3K27. In SU-DHL-10 mouse xeno- (IC50 ¼ 0.11 mM). In leukemia U937 and rhabdomyosarcoma RH4
graft models, dual therapy with GSK126 and romidepsin signifi- cancer cell lines, compound 36 was also an effective EZH2/HDAC
cantly delayed tumor growth and improved overall survival dual inhibitor by modulating known EZH2- and HDAC-dependent
compared with single-agent treatment [177]. Wang et al. reported histone markers, including H3K37Me3, Ac-H3, and Ac-tubulin.
that a combined treatment of either pan-HDAC inhibitor or a class I However, compound 36 did not show exciting antiproliferative
HDAC inhibitor with EZH2 inhibitors displayed a robust synergistic activity in several tested cancer cell lines, including U937, RH4,
effect in inducing histone H3K27 acetylation and target gene THP1, SH-N-SK, and U87, with IC50 values from 9 to 25 mM. Treating
expression, leading to rapid induction of apoptosis and stronger RH4 cancer cells with compound 36 induced apoptosis, caused cell
growth suppression of small cell carcinoma of the ovary and hy- cycle arrest in the subG1 phase, and promoted the expression of cell
percalcemic type cells. Concordant with the potent in vitro anti- surface adhesive receptors, CD11b and myogenin, markers of cell
cancer effects, synergetic in vivo efficacy between HDAC and EZH2 differentiation and myogenic differentiation, respectively. Also,
inhibitors were observed in the BIN67-xenograft models. Simulta- compound 36 upregulates E-cadherin expression without affecting
neous treatment of EPZ-6438 and Quisinostat completely inhibited the snail level, thereby contributing to the suppression of EMT
xenografted tumor growth and significantly prolonged the median process in U937 cells.
survival time. In contrast, single treatment with EPZ-6438 or Qui-
sinostat failed to or only mildly suppressed the tumor growth,
respectively [178]. Grinshtein and Co. found that a combination of
4.3. Dual inhibitors targeting LSD1 and G9a
EZH2 inhibitors (UNC1999, GSK126, and EPZ6438) and a unique
brain-penetrant class I HDAC inhibitor identified by Kim's group
In the process of pursuit of novel non-covalent LSD1 inhibitors
demonstrated strong synergism in two brain-tumor initiating cell
by Speranzini et al., several well-characterized G9a inhibitors
lines by enhancing apoptosis and increasing DNA damage [179]. In
featured with quinazoline skeleton were found to exhibit LSD1
TGBC2TKB gallbladder cancer cells, SAHA treatment down-
inhibitory activity. The representative compounds 37e40 with IC50
regulated the expression levels of HDAC1, HDAC2, and EZH2. This
values of 0.18, 0.18, 1.2, and 41.3 mM, respectively, toward G9a, are
led to the simultaneous activation of p16INK4a, E-cadherin, and p21
shown in Fig. 21 [182]. Compounds E11 and MC2694 showed the
and significant cell number decline. When combined with EZH2
highest binding affinity to LSD1 with thermal shifts of 6.5  C and
siRNA treatment, a synergistic antiproliferative effect was observed.
4.5  C, respectively. Their potent LSD1 inhibitory activity was
Interestingly, no such findings were obtained in human gallbladder
further confirmed by enzymatic and fluorescence polarization as-
epithelial cells, suggesting that the effect of SAHA depends on
says, showing Kd values of 243 nM and 385 nM, respectively. In
EZH2-mediated tumor suppressor loss.
MV4-11 leukemia cells, compound E11 did not induce any signifi-
cant increase in H3K4me2. However, qPCR analysis revealed that
4.2.2. Reported HDAC/EZH2 dual inhibitor E11 could strongly up-regulate the expression levels of Gfi1-B, an
Inspired by the above findings, Romanelli et al. developed a established target of LSD1, indicating the effective inhibition of
first-in-class dual HDAC/EZH2 inhibitor, 36, by fusing the structural intracellular LSD1. This potent dual LSD1/G9a inhibition of E11
features of SAHA through the “tail” of compound 35, a prototype of translated into promising antiproliferative effects on MV4-11 with
the pyridone-based EZH2 inhibitor reported by their group (Fig. 20) an IC50 value of 0.89 mM.
19
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 20. A pyridone-based HDAC/EZH2 dual inhibitor.

Fig. 21. Structure of LSD1/G9a dual inhibitors.

5. Dual inhibitor targeting histone writer and reader field, tested in phase Ib/IIa clinical trials for treating hematological
malignancies and advanced drug-resistant prostate cancer [183].
5.1. Crosstalk between BRD4 and CBP/p300 As important bromodomain-containing acetyllysine “readers,”
it is not surprising that an interplay exists between CBP/p300 and
Cyclic adenosine monophosphate-response element (CREB)- BRD4. Recent studies have confirmed that BRD4 and CBP/p300
binding protein (CBP) and adenoviral E1A binding protein of played an important role in regulating the well-known oncopro-
300 kDa (p300) are highly homologous lysine acetyltransferases. tein, c-Myc. Wu et al. found that in embryonic stem cells, BRD4
CBP and p300 proteins contain a single bromodomain adjacent to interacted with CBP/p300 through its two bromodomains to rein-
the histone acetyltransferase catalytic domain, thus functioning as force the histone acetyltransferase activity of CBP/p300, leading to
an acetyllysine “writer” and “reader.” As an acetyllysine “writer,” augmented acetylation of H3K27 and H3K56. Moreover, BRD4-
CBP/p300 can acetylate lysine residues on histone tails to remodel P300 interaction recruits chromatin remodeler, Brg1, to modulate
chromatin in a relaxed superstructure, and lysine residues on non- the chromatin structure, contributing to the transcription of
histone proteins to modulate their activities. Alternatively, as an pluripotency-associated genes, including Oct4 and Nanog in em-
acetyllysine “reader,” CBP/p300 binds to acetylated histones and bryonic stem cells (Fig. 22) [184]. In NMC driven by BRD4-NUT
non-histone proteins via their bromodomain, leading to the fusion oncoprotein, BRD4-NUT recruited and activated p300 via
recruitment of cellular transcriptional machinery to regulate gene the NUT portion to facilitate histone hyperacetylation and subse-
expression like c-Myc. Recent studies have demonstrated that quent BRD4-NUT recruitment to newly acetylated chromatin,
deregulation of CBP/p300 was closely related to the development, leading to uncontrolled spreading of BRD4-NUT across chromatin.
progression of various cancers, and tumor immunity. Given the Although BRD4 inhibitors showed limited efficacy against NMC due
vital role of CBP/p300 in driving cancer progression, depleting CBP/ to a narrow therapeutic window, combined treatment with CBP/
p300 levels exert potent therapeutic effects in cancers. CBP/p300 is p300 inhibitor, GNE-781, and BRD4 inhibitor, OTX015, coopera-
considered a potential anticancer target, and several CBP/p300 in- tively depleted c-Myc and synergistically inhibited NMC growth
hibitors targeting HAT domains or bromodomains have been re- [185].
ported recently. CCS1477 is the most advanced compound in this

20
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

Fig. 22. Molecular crosstalk model between BRD4 and P300. In embryonic stem cells, BRD4 partners with P300 and enhances its histone acetyltransferase activity. Augmenting
histone acetylation by BRD4-P300 interaction recruits the chromatin remodeler Brg1 to favor an open chromatin environment, thus faliticating the expression of pluripotency-
associated genes including Oct4 and Nanog.

5.2. Reported BRD4-CBP/p300 dual inhibitor in vitro and in vivo. In vitro, NEO2734 potently inhibited BRD4-
NUTþ NMC cells at low nM IC50 values and induced squamous
NEO2734 (compound 41) is a first-in-class and orally active dual differentiation. This observation is comparable with the clinical BET
epigenetic inhibitor, exhibiting a high affinity to BRD4 and CBP/ inhibitor, i-BET-762; however, disseminated BRD4-NUTþ NMC
p300 with Kd values ranging from 3.5 to 31 nM (Fig. 23) [186,187]. xenograft models treated with NEO2734 at a dose of 8 mg/kg
In vitro cell growth inhibition screening against a panel of 60 cell exhibited potent growth suppression and demonstrated markedly
lines derived from various cancer tissues was conducted by Ber- prolonged survival with median survival more than 100 d for PER-
toni's group. Results obtained demonstrated that NEO2734 403 mice and 44 d for 10e15 mice. This is more potent than i-BET-
exhibited promising antiproliferative activity in multiple cell lines, 762 with median survival of 41.5 d and 36.5 d, respectively [185].
especially in leukemia and lymphoma with median IC50 values of
280 nM and 300 nM, respectively, which are more potent than the 6. Discussion and conclusion
BRD4 inhibitor, iBET-762. In vivo, the antitumor efficacy of NEO2734
was evaluated in an ABC-DLBCL lymphoma xenograft cancer model Epigenetic dysregulation plays a crucial role in cancer devel-
at a dose of 10 mg/kg administered orally. NEO2734 significantly opment, progression, and acquisition of drug resistance. Epigenetic
induced tumor regression without apparent body weight loss, and modulators, especially histone epigenetic modulators, have
displayed higher efficacy than the BRD4 inhibitor, OXT-015, given at emerged as attractive therapeutic targets for cancer. After more
a higher dose of 50 mg/kg. Moreover, in an MV4-11 xenograft NOD/ than 20 years of unremitting pursuit, remarkable advances have
SCID mouse model, oral administration of NEO2734 even at a dose been made in this field, with eight epigenetic inhibitors approved
of 1 mg/kg for 14 d inhibited tumor growth. Treatment at a dose of for cancer treatment and numerous other candidates undergoing
3 mg/kg or 10 mg/kg enhanced tumor-repressive effect and was clinical trials. Despite recent significant processes achieved in
more potent than OXT015 at 10 mg/kg [187]. epigenetic therapy, to date, approved epigenetic inhibitors have
It has been reported that SPOP hotspot mutations, such as F133V mostly been confined to a subset of hematological cancers. Simul-
and W131R, disturbed its binding interactions with BRD4 proteins, taneously, their anticancer effects on solid tumors are unsatisfac-
resulting in decreased ubiquitination-mediated degradation and tory. Moreover, narrow safety margin, dose-limiting adverse
accumulation of BRD4 proteins, thus conferring high-level resis- events, and drug resistance limit their potential clinical use. New
tance to BRD4 inhibitors in prostate cancer. NEO2734 is active in strategies are urgently needed to overcome these challenges.
hotspot mutant (F133V) and non-hotspot mutant (Q165P) PCa cells Epigenetic modulators cooperatively or antagonistically regu-
in vitro and in vivo. NEO2734 induced marked suppression of Q165P late various histone modifications, creating a complicated network
and F133V C4-2 xenograft growth at a dose of 30 mg/kg, five days a for epigenetic transcription control during biological or pathogenic
week for three consecutive weeks. Also, NEO2734 showed strong development. Numerous combination therapies of two epigenetic
antitumor activity against 16 MM cell lines with a median IC50 value drugs with different action mechanisms are currently undergoing
of 140 nM. This is more potent than the single BRD4 inhibitors, exploration in preclinical or clinical trials to improve the effec-
iBET-762 and iBET-151, and the CBP/P300 inhibitor, CPI-637 [188]. tiveness of these single epigenetic target drugs in cancer therapy.
The anticancer efficacy of NEO2734 was also impressive in NMC Another attractive strategy for epigenetic drug development is the
discovery of dual inhibitors targeting different epigenetic modu-
lators. Compared with combination therapies, dual-target drugs
have several potential advantages, such as more predictable phar-
macokinetics, better patient compliance, reduced administration
dosage and toxicities. Excitingly, several dual epigenetic target in-
hibitors have been validated, and some of them showed superior
in vivo antitumor efficacy against solid tumors compared to the
combination of two individual single epigenetic target drugs.
Notably, the example of NEO2734 emphasizes the dual epigenetic
target inhibition as a promising strategy to overcome drug
resistance.
Despite their evident progress, development of dual epigenetic
Fig. 23. The structure of BRD4-CBP/p300 dual inhibitor, NEO2734. inhibitors remains a challenging task: the first critical challenge is
21
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

how to balance the potencies toward desired epigenetic modula- References


tors at an appropriate level to maximize the synergistic therapeutic
efficacy in vivo and safty profiles. Ideally, the in vitro activities for [1] T. Kouzarides, Chromatin modifications and their function, Cell 128 (2007)
693e705.
dual targets should be within an order of magnitude. However, in [2] L. Simo-Riudalbas, M. Esteller, Targeting the histone orthography of cancer:
most cases, it is not an easy task to achieve and potency against one drugs for writers, erasers and readers, Br. J. Pharmacol. 172 (2015)
target needs to be compromised. Moreover, differences in target 2716e2732.
[3] C.H. Arrowsmith, C. Bountra, P.V. Fish, K. Lee, M. Schapira, Epigenetic protein
occupancy, biodistribution of compounds and enzyme densities in families: a new frontier for drug discovery, Nat. Rev. Drug Discov. 11 (2012)
various tissues as well as time course of target inhibition pose 384e400.
roadblocks to develop dual epigenetic inhibitors with balanced [4] H.P. Mohammad, O. Barbash, C.L. Creasy, Targeting epigenetic modifications
in cancer therapy: erasing the roadmap to cancer, Nat. Med. 25 (2019)
in vivo activity [189]. 403e418.
Achieving satisfactory pharmacokinetic profiles without [5] A. Ganesan, P.B. Arimondo, M.G. Rots, C. Jeronimo, M. Berdasco, The timeline
compromising the activities of inhibitors will be another major of epigenetic drug discovery: from reality to dreams, Clin. Epigenet. 11
(2019) 174.
issue to solve. Most reported dual epigenetic inhibitors are
[6] S.J. Hogg, P.A. Beavis, M.A. Dawson, R.W. Johnstone, Targeting the epigenetic
designed using pharmacophore-linked or -fused strategy, which regulation of antitumour immunity, Nat. Rev. Drug Discov. 19 (2020)
generally results in high molecular weight and unfavorable phar- 776e800.
[7] S.E. Bates, D.L. Longo, Epigenetic therapies for cancer, N. Engl. J. Med. 383
macokinetic properties. Actually, some reported dual epigenetic
(2020) 650e663.
inhibitors do not show superior therapeutic benefits to traditional [8] Z. Liu, Y. Gao, X. Li, Cancer epigenetics and the potential of epigenetic drugs
epigenetic drugs, this is most likely due to the poor pharmacoki- for treating solid tumors, Expert. Rev. Anticancancer Ther. 19 (2018)
netics of obtained molecules. Apart from careful SAR explorations, 139e149.
[9] Y. Ishikawa, K. Nakayama, M. Morimoto, A. Mizutani, A. Nakayama,
pharmacophore-merged strategy and fragment-based screening K. Toyoshima, A. Hayashi, S. Takagi, R. Dairiki, H. Miyashita, S. Matsumoto,
strategy which give compounds with small molecular size need to K. Gamo, T. Nomura, K. Nakamura, Synergistic anti-AML effects of the LSD1
be fully explored to get dual epigenetic inhibitors with favorable inhibitor T-3775440 and the NEDD8-activating enzyme inhibitor pevonedi-
stat via transdifferentiation and DNA rereplication, Oncogenesis 6 (2017)
pharmacokinetics [190]. e377.
Systematic studies need to be conducted to ascertain the [10] S. Shu, C.Y. Lin, H.H. He, R.M. Witwicki, D.P. Tabassum, J.M. Roberts,
appropriate epigenetic target combinations that can achieve su- M. Janiszewska, S. Jin Huh, Y. Liang, J. Ryan, E. Doherty, H. Mohammed,
H. Guo, D.G. Stover, M.B. Ekram, G. Peluffo, J. Brown, C. D'Santos, I.E. Krop,
perior therapeutic efficacy through synergies. So far, most D. Dillon, M. McKeown, C. Ott, J. Qi, M. Ni, P.K. Rao, M. Duarte, S.-Y. Wu, C.-
described dual epigenetic inhibitors has particularly capitalized on M. Chiang, L. Anders, R.A. Young, E.P. Winer, A. Letai, W.T. Barry, J.S. Carroll,
the known HDAC inhibitors (especially alkyl hydroximic acid based H.W. Long, M. Brown, X. Shirley Liu, C.A. Meyer, J.E. Bradner, K. Polyak,
Response and resistance to BET bromodomain inhibitors in triple-negative
HDAC inhibitors) due to their proven anticancer efficacy, well-
breast cancer, Nature 529 (2016) 413e417.
studied SAR and high synthesis feasibility. However, the pharma- [11] A. Dhall, C.E. Weller, A. Chu, P.M.M. Shelton, C. Chatterjee, Chemically
cological liabilities of alkyl hydroximic acid scaffold often cause sumoylated histone H4 stimulates intranucleosomal demethylation by the
LSD1-CoREST complex, ACS Chem. Biol. 12 (2017) 2275e2280.
short pharmacokinetic half-lives, poor oral bioavailability and
[12] A.R. de Lera, A. Ganesan, Two-hit wonders: the expanding universe of
toxicity [191]. Although several dual epigenetic inhibitors that do multitargeting epigenetic agents, Curr. Opin. Chem. Biol. 57 (2020) 135e154.
not involve HDAC, such as NEO2734, have been reported, these [13] D. Tomaselli, A. Lucidi, D. Rotili, A. Mai, Epigenetic polypharmacology: a new
molecules are mainly discovered by serendipity. It is significant to frontier for epi-drug discovery, Med. Res. Rev. 40 (2019) 190e244.
[14] M.A. Glozak, N. Sengupta, X. Zhang, E. Seto, Acetylation and deacetylation of
explore effective strategies and techniques that can help efficiently non-histone proteins, Gene 363 (2005) 15e23.
design dual epigenetic inhibitors directed to diverse target com- [15] J.J. Kovacs, P.J.M. Murphy, S. Gaillard, X. Zhao, J.-T. Wu, C.V. Nicchitta,
binations. Epigenetic combination therapy results from clinical M. Yoshida, D.O. Toft, W.B. Pratt, T.-P. Yao, HDAC6 regulates Hsp90 acety-
lation and chaperone-dependent activation of glucocorticoid receptor, Mol.
observations or preclinical/preliminary explorations provide proof Cell. 18 (2005) 601e607.
of concepts for the rational design of dual epigenetic inhibitors. For [16] S. Ropero, M. Esteller, The role of histone deacetylases (HDACs) in human
example, BET inhibitor synergistic therapies with DOT1L inhibitor cancer, Mol. Oncol. 1 (2007) 19e25.
[17] C. Zagni, G. Floresta, G. Monciino, A. Rescifina, The search for potent, small-
or EZH2 inhibitor have achieved preliminary success in preclinical molecule HDACIs in cancer treatment: a decade after Vorinostat, Med. Res.
studies. Future attempts could use this information to pursue novel Rev. 37 (2017) 1373e1428.
dual epigenetic target inhibitors. [18] T.C.S. Ho, A.H.Y. Chan, A. Ganesan, Thirty years of HDAC inhibitors: 2020
insight and hindsight, J. Med. Chem. 63 (2020) 12460e12484.
Considering the tight connections between histone epigenetic
[19] V. Patil, Q.H. Sodji, J.R. Kornacki, M. Mrksich, A.K. Oyelere, 3-Hydroxypyridin-
modulators and the proven anticancer potency of reported dual 2-thione as novel zinc binding group for selective histone deacetylase in-
epigenetic inhibitors, the development of dual epigenetic target hibition, J. Med. Chem. 56 (2013) 3492e3506.
[20] F. Dehmel, S. Weinbrenner, H. Julius, T. Ciossek, T. Maier, T. Stengel, K. Fettis,
drugs will become a research hotspot. We foresee that the
C. Burkhardt, H. Wieland, T. Beckers, Trithiocarbonates as a novel class of
continuous deciphering of the epigenetic networks in cancer will HDAC inhibitors: SAR studies, isoenzyme selectivity, and pharmacological
further spark the discovery of dual epigenetic target inhibitors profiles, J. Med. Chem. 51 (2008) 3985e4001.
soon. [21] T. Liu, Y. Wan, Y. Xiao, C. Xia, G. Duan, Dual-target inhibitors based on
HDACs: novel antitumor agents for cancer therapy, J. Med. Chem. 63 (2020)
8977e9002.
Declaration of competing interest [22] Y. Luan, J. Li, J.A. Bernatchez, R. Li, Kinase and histone deacetylase hybrid
inhibitors for cancer therapy, J. Med. Chem. 62 (2019) 3171e3183.
[23] C. Dhalluin, J.E. Carlson, L. Zeng, C. He, A.K. Aggarwal, M.-M. Zhou, M.-
The authors declare that they have no known competing M. Zhou, Structure and ligand of a histone acetyltransferase bromodomain,
financial interests or personal relationships that could have Nature 399 (1999) 491e496.
appeared to influence the work reported in this paper. [24] P. Filippakopoulos, J. Qi, S. Picaud, Y. Shen, W.B. Smith, O. Fedorov,
E.M. Morse, T. Keates, T.T. Hickman, I. Felletar, M. Philpott, S. Munro,
M.R. McKeown, Y. Wang, A.L. Christie, N. West, M.J. Cameron, B. Schwartz,
Acknowledgments T.D. Heightman, N. La Thangue, C.A. French, O. Wiest, A.L. Kung, S. Knapp,
J.E. Bradner, Selective inhibition of BET bromodomains, Nature 468 (2010)
1067e1073.
This work was supported by the National Natural Science re, S. Rousseaux, U. Steuerwald, M. Soler-Lo  pez, S. Curtet, A.-
[25] J. Morinie
Foundation of China (Project No. U1704184); Science and Tech- L. Vitte, J. Govin, J. Gaucher, K. Sadoul, D.J. Hart, J. Krijgsveld, S. Khochbin,
nology Tackling Key Project of Henan Province (Project No. C.W. Müller, C. Petosa, Cooperative binding of two acetylation marks on a
histone tail by a single bromodomain, Nature 461 (2009) 664e668.
182102310214); Natural Science Foundation of Henan Province €sken, M. Geyer, Brd4 activates P-TEFb for
[26] F. Itzen, A.K. Greifenberg, C.A. Bo
(Project No. 202300410325).
22
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

RNA polymerase II CTD phosphorylation, Nucleic Acids Res. 42 (2014) acetylation and chromatin modification enhances BET inhibitor effects in B-
7577e7590. cell lymphoma cells, Sci. Rep. 9 (2019) 16495.
[27] B. Donati, E. Lorenzini, A. Ciarrocchi, BRD4 and Cancer: going beyond tran- [48] J.C. Enssle, C. Boedicker, M. Wanior, M. Vogler, S. Knapp, S. Fulda, Co-tar-
scriptional regulation, Mol. Canc. 17 (2018) 164. geting of BET proteins and HDACs as a novel approach to trigger apoptosis in
[28] B.N. Devaiah, B.A. Lewis, N. Cherman, M.C. Hewitt, B.K. Albrecht, P.G. Robey, rhabdomyosarcoma cells, Canc. Lett. 428 (2018) 160e172.
K. Ozato, R.J. Sims, D.S. Singer, BRD4 is an atypical kinase that phosphorylates [49] S. Liu, F. Li, L. Pan, Z. Yang, Y. Shu, W. Lv, P. Dong, W. Gong, BRD4 inhibitor
Serine2 of the RNA Polymerase II carboxy-terminal domain, Proc. Natl. Acad. and histone deacetylase inhibitor synergistically inhibit the proliferation of
Sci. U.S.A. 109 (2012) 6927e6932. gallbladder cancer in vitro and in vivo, Canc. Sci. 110 (2019) 2493e2506.
[29] O. Bechter, P. Scho € ffski, Make your best BET: the emerging role of BET in- [50] A.S. Holscher, W.A. Schulz, M. Pinkerneil, G. Niegisch, M.J. Hoffmann, Com-
hibitor treatment in malignant tumors, Pharmacol. Therapeut. 208 (2020) bined inhibition of BET proteins and class I HDACs synergistically induces
107479. apoptosis in urothelial carcinoma cell lines, Clin. Epigenet. 10 (2018) 1.
[30] M.E. White, J.M. Fenger, W.E. Carson, Emerging roles of and therapeutic [51] A. Heinemann, Wilmott, D. Gunatilake, C. Cullinane, R.D. Paoli-Iseppi,
strategies targeting BRD4 in cancer, Cell, Immunol. 337 (2019) 48e53. J. Madore, D. Strbenac, K. Gowrishankar, J.C. Tiffen, G.A. McArthur, P. Hersey,
[31] P. Tang, J. Zhang, J. Liu, C.M. Chiang, L. Ouyang, Targeting bromodomain and R.K. Prinjha, S.J. Gallagher, Combining BET and HDAC inhibitors synergisti-
extraterminal proteins for drug discovery: from current progress to tech- cally induces apoptosis of melanoma and suppresses AKT and YAP signaling,
nological development, J. Med. Chem. 64 (2021) 2419e2435. Oncotarget 6 (2015) 21507e21512.
[32] E. Kulikowski, B.D. Rakai, N.C.W. Wong, Inhibitors of bromodomain and [52] J. Bhadury, L.M. Nilsson, S.V. Muralidharan, L.C. Green, Z. Li, E.M. Gesner,
extra-terminal proteins for treating multiple human diseases, Med. Res. Rev. H.C. Hansen, U.B. Keller, K.G. McLure, J.A. Nilsson, BET and HDAC inhibitors
41 (2020) 223e245. induce similar genes and biological effects and synergize to kill in Myc-
[33] Z. Liu, P. Wang, H. Chen, E.A. Wold, B. Tian, A.R. Brasier, J. Zhou, Drug dis- induced murine lymphoma, Proc. Natl. Acad. Sci. U.S.A. 111 (2014)
covery targeting bromodomain-containing protein 4, J. Med. Chem. 60 2721e2730.
(2017) 4533e4558. [53] S. He, G. Dong, Y. Li, S. Wu, W. Wang, C. Sheng, Potent dual BET/HDAC in-
[34] S. Shu, H.-J. Wu, J.Y. Ge, R. Zeid, I.S. Harris, B. Jovanovic, K. Murphy, B. Wang, hibitors for efficient treatment of pancreatic cancer, Angew. Chem. Int. Ed.
X. Qiu, J.E. Endress, J. Reyes, K. Lim, A. Font-Tello, S. Syamala, T. Xiao, Engl. 59 (2020) 3028e3032.
C.S. Reddy Chilamakuri, E.K. Papachristou, C. D'Santos, J. Anand, K. Hinohara, [54] S. Laszig, C. Boedicker, T. Weiser, S. Knapp, S. Fulda, The novel dual BET/
W. Li, T.O. McDonald, A. Luoma, R.J. Modiste, Q.-D. Nguyen, B. Michel, HDAC inhibitor TW09 mediates cell death by mitochondrial apoptosis in
P. Cejas, C. Kadoch, J.D. Jaffe, K.W. Wucherpfennig, J. Qi, X.S. Liu, H. Long, rhabdomyosarcoma cells, Canc. Lett. 486 (2020) 46e57.
M. Brown, J.S. Carroll, J.S. Brugge, J. Bradner, F. Michor, K. Polyak, Synthetic [55] X. Zhang, T. Zegar, T. Weiser, F.H. Hamdan, B.T. Berger, R. Lucas,
lethal and resistance interactions with BET bromodomain inhibitors in D.I. Balourdas, S. Ladigan, P.F. Cheung, S.T. Liffers, M. Trajkovic-Arsic,
triple-negative breast cancer, Mol. Cell. 78 (2020) 1096e1113. B. Scheffler, A.C. Joerger, S.A. Hahn, S.A. Johnsen, S. Knapp, J.T. Siveke,
[35] P. Zhang, D. Wang, Y. Zhao, S. Ren, K. Gao, Z. Ye, S. Wang, C.-W. Pan, Y. Zhu, Characterization of a dual BET/HDAC inhibitor for treatment of pancreatic
Y. Yan, Y. Yang, D. Wu, Y. He, J. Zhang, D. Lu, X. Liu, L. Yu, S. Zhao, Y. Li, D. Lin, ductal adenocarcinoma, Int. J. Canc. 147 (2020) 2847e2861.
Y. Wang, L. Wang, Y. Chen, Y. Sun, C. Wang, H. Huang, Intrinsic BET inhibitor [56] Z. Pan, X. Li, Y. Wang, Q. Jiang, L. Jiang, M. Zhang, N. Zhang, F. Wu, B. Liu,
resistance in SPOP-mutated prostate cancer is mediated by BET protein G. He, Discovery of thieno[2,3-d]pyrimidine-based hydroxamic acid de-
stabilization and AKTemTORC1 activation, Nat. Med. 23 (2017) 1055e1062. rivatives as bromodomain-containing protein 4/histone deacetylase dual
[36] J. Fu, H.-G. Yoon, J. Qin, J. Wong, Regulation of P-TEFb elongation complex inhibitors induce autophagic cell death in colorectal carcinoma cells, J. Med.
activity by CDK9 acetylation, Mol. Cell Biol. 27 (2007) 4641e4651. Chem. 63 (2020) 3678e3700.
[37] S. Pinz, S. Unser, D. Buob, P. Fischer, B. Jobst, A. Rascle, Deacetylase inhibitors [57] E.J. Faivre, K.F. McDaniel, D.H. Albert, S.R. Mantena, J.P. Plotnik, D. Wilcox,
repress STAT5-mediated transcription by interfering with bromodomain and L. Zhang, M.H. Bui, G.S. Sheppard, L. Wang, V. Sehgal, X. Lin, X. Huang, X. Lu,
extra-terminal (BET) protein function, Nucleic Acids Res. 43 (2015) T. Uziel, P. Hessler, L.T. Lam, R.J. Bellin, G. Mehta, S. Fidanze, J.K. Pratt, D. Liu,
3524e3545. L.A. Hasvold, C. Sun, S.C. Panchal, J.J. Nicolette, S.L. Fossey, C.H. Park,
[38] X. Hu, X. Lu, R. Liu, N. Ai, Z. Cao, Y. Li, J. Liu, B. Yu, K. Liu, H. Wang, C. Zhou, K. Longenecker, L. Bigelow, M. Torrent, S.H. Rosenberg, W.M. Kati, Y. Shen,
Y. Wang, A. Han, F. Ding, R. Chen, Histone cross-talk connects protein Selective inhibition of the BD2 bromodomain of BET proteins in prostate
phosphatase 1a (PP1a) and histone deacetylase (HDAC) pathways to regulate cancer, Nature 578 (2020) 306e310.
the functional transition of bromodomain-containing 4 (BRD4) for inducible [58] G.S. Sheppard, L. Wang, S.D. Fidanze, L.A. Hasvold, D. Liu, J.K. Pratt, C.H. Park,
gene expression, J. Biol. Chem. 289 (2014) 23154e23167. K. Longenecker, W. Qiu, M. Torrent, P.J. Kovar, M. Bui, E. Faivre, X. Huang,
[39] V.K. Mishra, F. Wegwitz, R.L. Kosinsky, M. Sen, R. Baumgartner, T. Wulff, X. Lin, D. Wilcox, L. Zhang, Y. Shen, D.H. Albert, T.J. Magoc, G. Rajaraman,
J.T. Siveke, H.U. Schildhaus, Z. Najafova, V. Kari, H. Kohlhof, E. Hessmann, W.M. Kati, K.F. McDaniel, Discovery of N-Ethyl-4-[2-(4-fluoro-2,6-dimethyl-
S.A. Johnsen, Histone deacetylase class-I inhibition promotes epithelial gene phenoxy)-5-(1-hydroxy-1-methyl-ethyl)phenyl]-6-methyl-7-oxo-1H-pyr-
expression in pancreatic cancer cells in a BRD4- and MYC-dependent rolo[2,3-c]pyridine-2-carboxamide (ABBV-744), a BET bromodomain inhib-
manner, Nucleic Acids Res. 45 (2017) 6334e6349. itor with selectivity for the second bromodomain, J. Med. Chem. 63 (2020)
[40] W. Fiskus, S. Sharma, J. Qi, J.A. Valenta, L.J. Schaub, B. Shah, K. Peth, 5585e5623.
B.P. Portier, M. Rodriguez, S.G. Devaraj, M. Zhan, J. Sheng, S.P. Iyer, [59] J. Chen, Y. Li, J. Zhang, M. Zhang, A. Wei, H. Liu, Z. Xie, W. Ren, W. Duan,
J.E. Bradner, K.N. Bhalla, Highly active combination of BRD4 antagonist and Z. Zhang, A. Shen, Y. Hu, Discovery of selective HDAC/BRD4 dual inhibitors as
histone deacetylase inhibitor against human acute myelogenous leukemia epigenetic probes, Eur. J. Med. Chem. 209 (2021) 112868.
cells, Mol. Canc. Therapeut. 13 (2014) 1142e1154. [60] D.S. Hewings, O. Fedorov, P. Filippakopoulos, S. Martin, S. Picaud, A. Tumber,
[41] P.K. Mazur, A. Herner, S.S. Mello, M. Wirth, S. Hausmann, F.J. Sanchez-Rivera, C. Wells, M.M. Olcina, K. Freeman, A. Gill, A.J. Ritchie, D.W. Sheppard,
S.M. Lofgren, T. Kuschma, S.A. Hahn, D. Vangala, M. Trajkovic-Arsic, A. Gupta, A.J. Russell, E.M. Hammond, S. Knapp, P.E. Brennan, S.J. Conway, Optimiza-
I. Heid, P.B. Noel, R. Braren, M. Erkan, J. Kleeff, B. Sipos, L.C. Sayles, tion of 3,5-dimethylisoxazole derivatives as potent bromodomain ligands,
M. Heikenwalder, E. Hessmann, V. Ellenrieder, I. Esposito, T. Jacks, J. Med. Chem. 56 (2013) 3217e3227.
J.E. Bradner, P. Khatri, E.A. Sweet-Cordero, L.D. Attardi, R.M. Schmid, [61] Z. Zhang, S. Hou, H. Chen, T. Ran, F. Jiang, Y. Bian, D. Zhang, Y. Zhi, L. Wang,
G. Schneider, J. Sage, J.T. Siveke, Combined inhibition of BET family proteins L. Zhang, H. Li, Y. Zhang, W. Tang, T. Lu, Y. Chen, Targeting epigenetic reader
and histone deacetylases as a potential epigenetics-based therapy for and eraser: rational design, synthesis and in vitro evaluation of dimethyli-
pancreatic ductal adenocarcinoma, Nat. Med. 21 (2015) 1163e1171. soxazoles derivatives as BRD4/HDAC dual inhibitors, Bioorg. Med. Chem. Lett
[42] H. Zeng, J. Qu, N. Jin, J. Xu, C. Lin, Y. Chen, X. Yang, X. He, S. Tang, X. Lan, 26 (2016) 2931e2935.
X. Yang, Z. Chen, M. Huang, J. Ding, M. Geng, Feedback activation of leukemia [62] G. Cheng, Z. Wang, J. Yang, Y. Bao, Q. Xu, L. Zhao, D. Liu, Design, synthesis and
inhibitory factor receptor limits response to histone deacetylase inhibitors in biological evaluation of novel indole derivatives as potential HDAC/BRD4
breast cancer, Canc. Cell 30 (2016) 459e473. dual inhibitors and anti-leukemia agents, Bioorg. Chem. 84 (2019) 410e417.
[43] G. Borbely, L.-A. Haldosen, K. Dahlman-Wright, C. Zhao, Induction of USP17 [63] S. Amemiya, T. Yamaguchi, Y. Hashimoto, T. Noguchi-Yachide, Synthesis and
by combining BET and HDAC inhibitors in breast cancer cells, Oncotarget 6 evaluation of novel dual BRD4/HDAC inhibitors, Bioorg. Med. Chem. 25
(2015) 33623e33635. (2017) 3677e3684.
[44] Y. Zhang, C.T. Ishida, W. Ishida, S.-F.L. Lo, J. Zhao, C. Shu, E. Bianchetti, [64] M. Shao, L. He, L. Zheng, L. Huang, Y. Zhou, T. Wang, Y. Chen, M. Shen,
G. Kleiner, M.J. Sanchez-Quintero, C.M. Quinzii, M.-A. Westhoff, G. Karpel- F. Wang, Z. Yang, L. Chen, Structure-based design, synthesis and in vitro
Massler, P. Canoll, M.D. Siegelin, Combined HDAC and bromodomain protein antiproliferative effects studies of novel dual BRD4/HDAC inhibitors, Bioorg.
inhibition reprograms tumor cell metabolism and elicits synthetic lethality Med. Chem. Lett 27 (2017) 4051e4055.
in glioblastoma, Clin. Canc. Res. 24 (2018) 3941e3954. [65] ^ te
B.J. Klein, K.L. Cox, S.M. Jang, J. Co , M.G. Poirier, T.G. Kutateladze, Molecular
[45] W. Meng, B. Wang, W. Mao, J. Wang, Y. Zhao, Q. Li, C. Zhang, Y. Tang, J. Ma, basis for the PZP domain of BRPF1 association with chromatin, Structure 28
Enhanced efficacy of histone deacetylase inhibitor combined with bromo- (2020) 105e110.
domain inhibitor in glioblastoma, J. Exp. Clin. Canc. Res. 37 (2018) 241. [66] J. Zhu, C. Zhou, A. Caflisch, Structure-based discovery of selective BRPF1
[46] L. Zhao, J.P. Okhovat, E.K. Hong, Y.H. Kim, G.S. Wood, Preclinical studies bromodomain inhibitors, Eur. J. Med. Chem. 155 (2018) 337e352.
support combined inhibition of BET family proteins and histone deacetylases [67] N. Igoe, E.D. Bayle, O. Fedorov, C. Tallant, P. Savitsky, C. Rogers, D.R. Owen,
as epigenetic therapy for cutaneous T-cell lymphoma, Neoplasia 21 (2019) G. Deb, T.C. Somervaille, D.M. Andrews, N. Jones, A. Cheasty, H. Ryder,
82e92. P.E. Brennan, S. Muller, S. Knapp, P.V. Fish, Design of a biased potent small
[47] M.G. Cortiguera, L. García-Gaipo, S.D. Wagner, J. Leo n, A. Batlle-Lo pez, molecule inhibitor of the bromodomain and PHD finger-containing (BRPF)
M.D. Delgado, Suppression of BCL6 function by HDAC inhibitor mediated proteins suitable for cellular and in vivo studies, J. Med. Chem. 60 (2017)

23
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

668e680. M.R. Kaadige, S. Srivastava, S. Daniel Ampanattu, R. Rodriguez del Villar,


[68] T. Heimburg, F.R. Kolbinger, P. Zeyen, E. Ghazy, D. Herp, K. Schmidtkunz, J. Lang, H. Vankayalapati, B. Weissman, J.M. Trent, W.P.D. Hendricks,
J. Melesina, T.B. Shaik, F. Erdmann, M. Schmidt, C. Romier, D. Robaa, O. Witt, S. Sharma, The novel reversible LSD1 inhibitor SP-2577 promotes anti-tumor
I. Oehme, M. Jung, W. Sippl, Structure-based design and biological charac- immunity in SWItch/Sucrose-NonFermentable (SWI/SNF) complex mutated
terization of selective histone deacetylase 8 (HDAC8) inhibitors with anti- ovarian cancer, PloS One 15 (2020) e0235705.
neuroblastoma activity, J. Med. Chem. 60 (2017) 10188e10204. [90] Y. Fang, G. Liao, B. Yu, LSD1/KDM1A inhibitors in clinical trials: advances and
[69] E. Ghazy, P. Zeyen, D. Herp, M. Hügle, K. Schmidtkunz, F. Erdmann, D. Robaa, prospects, J. Hematol. Oncol. 12 (2019) 129.
M. Schmidt, E.R. Morales, C. Romier, S. Günther, M. Jung, W. Sippl, Design, [91] X.J. Dai, Y. Liu, X.P. Xiong, L.P. Xue, Y.C. Zheng, H.M. Liu, Tranylcypromine
synthesis, and biological evaluation of dual targeting inhibitors of histone based lysine-specific demethylase 1 inhibitor: summary and perspective,
deacetylase 6/8 and bromodomain BRPF1, Eur. J. Med. Chem. 200 (2020) J. Med. Chem. 63 (2020) 14197e14215.
112338. [92] S.A. Kim, J. Zhu, N. Yennawar, P. Eek, S. Tan, Crystal structure of the LSD1/
[70] Y. Shi, F. Lan, C. Matson, P. Mulligan, J.R. Whetstine, P.A. Cole, R.A. Casero, CoREST histone demethylase bound to its nucleosome substrate, Mol. Cell.
Y. Shi, Histone demethylation mediated by the nuclear amine oxidase ho- 78 (2020) 903e914.
molog LSD1, Cell 119 (2004) 941e953. [93] Y. Song, L. Dagil, L. Fairall, N. Robertson, M. Wu, T.J. Ragan, C.G. Savva,
[71] J. Huang, R. Sengupta, A.B. Espejo, M.G. Lee, J.A. Dorsey, M. Richter, S. Opravil, A. Saleh, N. Morone, M.B.A. Kunze, A.G. Jamieson, P.A. Cole, D.F. Hansen,
R. Shiekhattar, M.T. Bedford, T. Jenuwein, S.L. Berger, p53 is regulated by the J.W.R. Schwabe, Mechanism of crosstalk between the LSD1 demethylase and
lysine demethylase LSD1, Nature 449 (2007) 105e108. HDAC1 deacetylase in the CoREST complex, Cell Rep. 30 (2020) 2699e2711.
[72] J. Wang, S. Hevi, J.K. Kurash, H. Lei, F. Gay, J. Bajko, H. Su, W. Sun, H. Chang, [94] Y. Yang, W. Huang, R. Qiu, R. Liu, Y. Zeng, J. Gao, Y. Zheng, Y. Hou, S. Wang,
G. Xu, F. Gaudet, E. Li, T. Chen, The lysine demethylase LSD1 (KDM1) is W. Yu, S. Leng, D. Feng, Y. Wang, Z. Shen, LSD1 coordinates with the SIN3A/
required for maintenance of global DNA methylation, Nat. Genet. 41 (2009) HDAC complex and maintains sensitivity to chemotherapy in breast cancer,
125e129. J. Mol. Cell Biol. 10 (2018) 285e301.
[73] H. Kontaki, I. Talianidis, Lysine methylation regulates E2F1-induced cell [95] D.A. Nalawansha, M.K.H. Pflum, LSD1 substrate binding and gene expression
death, Mol. Cell. 39 (2010) 152e160. are affected by HDAC1-mediated deacetylation, ACS Chem. Biol. 12 (2016)
[74] H.S. Cho, T. Suzuki, N. Dohmae, S. Hayami, M. Unoki, M. Yoshimatsu, 254e264.
G. Toyokawa, M. Takawa, T. Chen, J.K. Kurash, H.I. Field, B.A. Ponder, [96] C. Cao, S.N. Vasilatos, R. Bhargava, J.L. Fine, S. Oesterreich, N.E. Davidson,
Y. Nakamura, R. Hamamoto, Demethylation of RB regulator MYPT1 by his- Y. Huang, Functional interaction of histone deacetylase 5 (HDAC5) and
tone demethylase LSD1 promotes cell cycle progression in cancer cells, Canc. lysine-specific demethylase 1 (LSD1) promotes breast cancer progression,
Res. 71 (2011) 655e660. Oncogene 36 (2017) 133e145.
[75] D. Willmann, S. Lim, S. Wetzel, E. Metzger, A. Jandausch, W. Wilk, M. Jung, [97] C. Cao, H. Wu, S.N. Vasilatos, U. Chandran, Y. Qin, Y. Wan, S. Oesterreich,
I. Forne, A. Imhof, A. Janzer, J. Kirfel, H. Waldmann, R. Schule, R. Buettner, N.E. Davidson, Y. Huang, HDAC5eLSD1 axis regulates antineoplastic effect of
Impairment of prostate cancer cell growth by a selective and reversible natural HDAC inhibitor sulforaphane in human breast cancer cells, Int. J.
lysine-specific demethylase 1 inhibitor, Int. J. Canc. 131 (2012) 2704e2709. Canc. 143 (2018) 1388e1401.
[76] H.P. Mohammad, K.N. Smitheman, C.D. Kamat, D. Soong, K.E. Federowicz, [98] Y. Huang, S.N. Vasilatos, L. Boric, P.G. Shaw, N.E. Davidson, Inhibitors of
G.S. Van Aller, J.L. Schneck, J.D. Carson, Y. Liu, M. Butticello, W.G. Bonnette, histone demethylation and histone deacetylation cooperate in regulating
S.A. Gorman, Y. Degenhardt, Y. Bai, M.T. McCabe, M.B. Pappalardi, J. Kasparec, gene expression and inhibiting growth in human breast cancer cells, Breast
X. Tian, K.C. McNulty, M. Rouse, P. McDevitt, T. Ho, M. Crouthamel, T.K. Hart, Canc. Res. Treat. 131 (2012) 777e789.
N.O. Concha, C.F. McHugh, W.H. Miller, D. Dhanak, P.J. Tummino, [99] W. Fiskus, S. Sharma, B. Shah, B.P. Portier, S.G. Devaraj, K. Liu, S.P. Iyer,
C.L. Carpenter, N.W. Johnson, C.L. Hann, R.G. Kruger, A DNA hypomethylation D. Bearss, K.N. Bhalla, Highly effective combination of LSD1 (KDM1A)
signature predicts antitumor activity of LSD1 inhibitors in SCLC, Canc. Cell 28 antagonist and pan-histone deacetylase inhibitor against human AML cells,
(2015) 57e69. Leukemia 28 (2014) 2155e2164.
[77] J. Ding, Z.M. Zhang, Y. Xia, G.Q. Liao, Y. Pan, S. Liu, Y. Zhang, Z.S. Yan, LSD1- [100] D. Welch, E. Kahen, B. Fridley, A.S. Brohl, C.L. Cubitt, D.R. Reed, Small
mediated epigenetic modification contributes to proliferation and metastasis molecule inhibition of lysine-specific demethylase 1 (LSD1) and histone
of colon cancer, Br. J. Canc. 109 (2013) 994e1003. deacetylase (HDAC) alone and in combination in Ewing sarcoma cell lines,
[78] Y. Wang, H. Zhang, Y. Chen, Y. Sun, F. Yang, W. Yu, J. Liang, L. Sun, X. Yang, PloS One 14 (2019) e0222228.
L. Shi, R. Li, Y. Li, Y. Zhang, Q. Li, X. Yi, Y. Shang, LSD1 is a subunit of the NuRD [101] S.N. Vasilatos, T.A. Katz, S. Oesterreich, Y. Wan, N.E. Davidson, Y. Huang,
complex and targets the metastasis programs in breast cancer, Cell 138 Crosstalk between lysine-specific demethylase 1 (LSD1) and histone deace-
(2009) 660e672. tylases mediates antineoplastic efficacy of HDAC inhibitors in human breast
[79] S. Zhang, M. Liu, Y. Yao, B. Yu, H. Liu, Targeting LSD1 for acute myeloid cancer cells, Carcinogenesis 34 (2013) 1196e1207.
leukemia (AML) treatment, Pharmacol. Res. 164 (2021) 105335. [102] T. Haydn, E. Metzger, R. Schuele, S. Fulda, Concomitant epigenetic targeting
[80] M.R. Pandey, E.S. Wang, What potential is there for LSD1 inhibitors to reach of LSD1 and HDAC synergistically induces mitochondrial apoptosis in
approval for AML? Expet Opin. Emerg. Drugs 24 (2019) 205e212. rhabdomyosarcoma cells, Cell Death Dis. 8 (2017) e2879.
[81] T. Lin, A. Ponn, X. Hu, B.K. Law, J. Lu, Requirement of the histone demethylase [103] M.M. Singh, B. Johnson, A. Venkatarayan, E.R. Flores, J. Zhang, X. Su,
LSD1 in Snai1-mediated transcriptional repression during epithelial- M. Barton, F. Lang, J. Chandra, Preclinical activity of combined HDAC and
mesenchymal transition, Oncogene 29 (2010) 4896e4904. KDM1A inhibition in glioblastoma, Neuro Oncol. 17 (2015) 1463e1473.
[82] W.A. Whyte, S. Bilodeau, D.A. Orlando, H.A. Hoke, G.M. Frampton, C.T. Foster, [104] M.M. Singh, C.A. Manton, K.P. Bhat, W.W. Tsai, K. Aldape, M.C. Barton,
S.M. Cowley, R.A. Young, Enhancer decommissioning by LSD1 during em- J. Chandra, Inhibition of LSD1 sensitizes glioblastoma cells to histone
bryonic stem cell differentiation, Nature 482 (2012) 221e225. deacetylase inhibitors, Neuro Oncol. 13 (2011) 894e903.
[83] S. Ambrosio, A. Ballabio, B. Majello, Histone methyl-transferases and [105] J.H. Kalin, M. Wu, A.V. Gomez, Y. Song, J. Das, D. Hayward, N. Adejola, M. Wu,
demethylases in the autophagy regulatory network: the emerging role of I. Panova, H.J. Chung, E. Kim, H.J. Roberts, J.M. Roberts, P. Prusevich,
KDM1A/LSD1 demethylase, Autophagy 15 (2018) 187e196. J.R. Jeliazkov, S.S. Roy Burman, L. Fairall, C. Milano, A. Eroglu, C.M. Proby,
[84] W. Sheng, M.W. LaFleur, T.H. Nguyen, S. Chen, A. Chakravarthy, J.R. Conway, A.T. Dinkova-Kostova, W.W. Hancock, J.J. Gray, J.E. Bradner, S. Valente,
Y. Li, H. Chen, H. Yang, P.H. Hsu, E.M. Van Allen, G.J. Freeman, D.D. De Car- A. Mai, N.M. Anders, M.A. Rudek, Y. Hu, B. Ryu, J.W.R. Schwabe, A. Mattevi,
valho, H.H. He, A.H. Sharpe, Y. Shi, LSD1 ablation stimulates anti-tumor R.M. Alani, P.A. Cole, Targeting the CoREST complex with dual histone
immunity and enables checkpoint blockade, Cell 174 (2018) 549e563 e519. deacetylase and demethylase inhibitors, Nat. Commun. 9 (2018) 53.
[85] T. Maes, C. Molinero, R.M. Antonijoan, J.M. Ferrero-Cafiero, J. Martínez- [106] J.N. Anastas, B.M. Zee, J.H. Kalin, M. Kim, R. Guo, S. Alexandrescu, M.A. Blanco,
Colomer, C. Mascaro, M.I. Arevalo, First-in-Human phase I results show S. Giera, S.M. Gillespie, J. Das, M. Wu, S. Nocco, D.M. Bonal, Q.D. Nguyen,
safety, tolerability and brain penetrance of Ory-2001, an epigenetic drug M.L. Suva, B.E. Bernstein, R. Alani, T.R. Golub, P.A. Cole, M.G. Filbin, Y. Shi, Re-
targeting Lsd1 and Mao-B, Alzheimer's Dementia 13 (2017) 1573e1574. programing chromatin with a bifunctional LSD1/HDAC inhibitor induces
[86] S.H. Lee, M. Stubbs, X.M. Liu, M. Diamond, V. Dostalik, M. Ye, Y. Lo, M. Favata, therapeutic differentiation in DIPG, Canc. Cell 36 (2019) 1e17.
G. Yang, K. Gallagher, L. Leffet, C. He, L. Wu, A. Margulis, M. Covington, [107] Y.C. Duan, Y.C. Ma, W.P. Qin, L.N. Ding, Y.C. Zheng, Y.L. Zhu, X.Y. Zhai, J. Yang,
R. Wynn, W. Yao, G. Hollis, R. Huber, B. Ruggeri, P. Scherle, Abstract 4712: C.Y. Ma, Y.Y. Guan, Design and synthesis of tranylcypromine derivatives as
discovery of INCB059872, a novel FAD-directed LSD1 inhibitor that is novel LSD1/HDACs dual inhibitors for cancer treatment, Eur. J. Med. Chem.
effective in preclinical models of human and murine AML, Canc. Res. 76 140 (2017) 392e402.
(2016) 4712. [108] M Naveen Sadhu, Dhanalakshmi Sivanandhan, Chandru Gajendran,
[87] G. Civenni, G. Zoppi, R. Vazquez, D. Shinde, A. Paganoni, A. Kokanovic, Subramanyam Tantry, Purushottam Dewang, Murugan Kannan,
S.H. Lee, B. Ruggeri, G.M. Carbone, C.V. Catapano, Abstract 1379: Srinatha Chickamunivenkatappa, Mohd Zainuddin, Sreekala Nair,
INCB059872, a novel FAD-directed LSD1 Inhibitor, is active in prostate cancer S. Krishnakumar Vaithilingam, Rajagopal, Novel dual LSD1/HDAC6 inhibitors
models and impacts prostate cancer stem-like cells, Canc. Res. 78 (2018) for the treatment of multiple myelom, Bioorg. Med. Chem. Lett 34 (2021)
1379. 127763.
[88] T. Kanouni, C. Severin, R.W. Cho, N.Y.Y. Yuen, J. Xu, L. Shi, C. Lai, J.R. Del [109] A. Milelli, C. Marchetti, E. Turrini, E. Catanzaro, R. Mazzone, D. Tomaselli,
Rosario, R.K. Stansfield, L.N. Lawton, D. Hosfield, S. O'Connell, M.M. Kreilein, C. Fimognari, V. Tumiatti, A. Minarini, Novel polyamine-based Histone
P. Tavares-Greco, Z. Nie, S.W. Kaldor, J.M. Veal, J.A. Stafford, Y.K. Chen, Dis- deacetylases-Lysine demethylase 1 dual binding inhibitors, Bioorg. Med.
covery of CC-90011: a potent and selective reversible inhibitor of lysine Chem. Lett 28 (2018) 1001e1004.
specific demethylase 1 (LSD1), J. Med. Chem. 63 (2020) 14522e14529. [110] M. Pinkerneil, M.J. Hoffmann, H. Kohlhof, W.A. Schulz, G. Niegisch, Evalua-
[89] S. Saladi, R. Soldi, T. Ghosh Halder, A. Weston, T. Thode, K. Drenner, R. Lewis, tion of the therapeutic potential of the novel isotype specific HDAC inhibitor

24
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

4SC-202 in urothelial carcinoma cell lines, Targeted Oncol. 11 (2016) 2027.


783e798. [133] M. Tachibana, K. Sugimoto, T. Fukushima, Y. Shinkai, SET domain-containing
[111] K. Inui, Z. Zhao, J. Yuan, S. Jayaprakash, L.T.M. Le, S. Drakulic, B. Sander, protein, G9a, is a novel lysine-preferring mammalian histone methyl-
M.M. Golas, Stepwise assembly of functional C-terminal REST/NRSF tran- transferase with hyperactivity and specific selectivity to lysines 9 and 27 of
scriptional repressor complexes as a drug target, Protein Sci. 26 (2017) histone H3, J. Biol. Chem. 276 (2001) 25309e25317.
997e1011. [134] A.J. Bannister, P. Zegerman, J.F. Partridge, E.A. Miska, J.O. Thomas,
[112] M. Fu, F. Wan, Z. Li, F. Zhang, 4SC-202 activates ASK1-dependent mito- R.C. Allshire, T. Kouzarides, Selective recognition of methylated lysine 9 on
chondrial apoptosis pathway to inhibit hepatocellular carcinoma cells, Bio- histone H3 by the HP1 chromo domain, Nature 410 (2001) 120e124.
chem. Biophys. Res. Commun. 471 (2016) 267e273. [135] P. Rathert, A. Dhayalan, M. Murakami, X. Zhang, R. Tamas, R. Jurkowska,
[113] S.M. Messerli, M.M. Hoffman, E.Z. Gnimpieba, H. Kohlhof, R.D. Bhardwaj, Y. Komatsu, Y. Shinkai, X. Cheng, A. Jeltsch, Protein lysine methyltransferase
4SC-202 as a potential treatment for the pediatric brain tumor medullo- G9a acts on non-histone targets, Nat. Chem. Biol. 4 (2008) 344e346.
blastoma, Brain Sci. 7 (2017). [136] A. Chopra, W.C. Cho, W.G. Willmore, K.K. Biggar, Hypoxia-inducible lysine
[114] H. Zhijun, W. Shusheng, M. Han, L. Jianping, Q. Li-sen, L. Dechun, Pre-clinical methyltransferases: G9a and GLP hypoxic regulation, non-histone substrate
characterization of 4SC-202, a novel class I HDAC inhibitor, against colorectal modification, and pathological relevance, Front. Genet. 11 (2020) 579636.
cancer cells, Tumor, Biol. 37 (2016) 10257e10267. [137] J. Huang, J. Dorsey, S. Chuikov, X. Zhang, T. Jenuwein, D. Reinberg, S.L. Berger,
[115] M. Wobser, A. Weber, A. Glunz, S. Tauch, K. Seitz, T. Butelmann, S. Hesbacher, G9a and Glp methylate lysine 373 in the tumor suppressor p53, J. Biol. Chem.
M. Goebeler, R. Bartz, H. Kohlhof, D. Schrama, R. Houben, Elucidating the 285 (2010) 9636e9641.
mechanism of action of domatinostat (4SC-202) in cutaneous T cell lym- [138] J.S. Lee, Y. Kim, I.S. Kim, B. Kim, H.J. Choi, J.M. Lee, H.-J.R. Shin, J.H. Kim, J.-
phoma cells, J. Hematol. Oncol. 12 (2019) 30. Y. Kim, S.-B. Seo, H. Lee, O. Binda, O. Gozani, G.L. Semenza, M. Kim, K.I. Kim,
[116] A.C. Bretz, U. Parnitzke, K. Kronthaler, T. Dreker, R. Bartz, F. Hermann, D. Hwang, S.H. Baek, Negative regulation of hypoxic responses via induced
A. Ammendola, T. Wulff, S. Hamm, Domatinostat favors the immunotherapy reptin methylation, Mol. Cell. 39 (2010) 71e85.
response by modulating the tumor immune microenvironment (TIME), [139] M. Tachibana, G9a histone methyltransferase plays a dominant role in
J. Immunother. Canc. 7 (2019) 294. euchromatic histone H3 lysine 9 methylation and is essential for early
[117] Y. Duan, W. Qin, F. Suo, X. Zhai, Y. Guan, X. Wang, Y. Zheng, H. Liu, Design, embryogenesis, Gene Dev. 16 (2002) 1779e1791.
synthesis and in vitro evaluation of stilbene derivatives as novel LSD1 in- [140] Q. Yang, Q. Zhu, X. Lu, Y. Du, L. Cao, C. Shen, T. Hou, M. Li, Z. Li, C. Liu, D. Wu,
hibitors for AML therapy, Bioorg. Med. Chem. 26 (2018) 6000e6014. X. Xu, L. Wang, H. Wang, Y. Zhao, Y. Yang, W.-G. Zhu, G9a coordinates with
[118] Y.C. Duan, L.F. Jin, H.M. Ren, S.J. Zhang, Y.J. Liu, Y.T. Xu, Z.H. He, Y. Song, the RPA complex to promote DNA damage repair and cell survival, Proc. Natl.
H. Yuan, S.-H. Chen, Y.Y. Guan, Design, synthesis, and biological evaluation of Acad. Sci. U.S.A. 114 (2017) E6054eE6063.
novel dual inhibitors targeting lysine specific demethylase 1 (LSD1) and [141] C. Yin, X. Ke, R. Zhang, J. Hou, Z. Dong, F. Wang, K. Zhang, X. Zhong, L. Yang,
histone deacetylases (HDAC) for treatment of gastric cancer, Eur. J. Med. H. Cui, G9a promotes cell proliferation and suppresses autophagy in gastric
Chem. 220 (2021) 113453. cancer by directly activating mTOR, FASEB. J. 33 (2019) 14036e14050.
[119] R.J. Klose, E.M. Kallin, Y. Zhang, JmjC-domain-containing proteins and his- [142] F. Casciello, K. Windloch, F. Gannon, J.S. Lee, Functional role of G9a histone
tone demethylation, Nat. Rev. Genet. 7 (2006) 715e727. methyltransferase in cancer, Front. Immunol. 6 (2015) 487.
[120] L.J. Walport, R.J. Hopkinson, C.J. Schofield, Mechanisms of human histone and [143] H. Wang, L. Cui, D. Li, M. Fan, Z. Liu, C. Liu, S. Pan, L. Zhang, H. Zhang, Y. Zhao,
nucleic acid demethylases, Curr. Opin. Chem. Biol. 16 (2012) 525e534. Overexpression of PSAT1 regulated by G9A sustains cell proliferation in
[121] G.J. Yang, M.H. Zhu, X.J. Lu, Y.J. Liu, J.F. Lu, C.H. Leung, D.L. Ma, J. Chen, The colorectal cancer, Signal Transduct. Targeted Ther. 5 (2020) 47.
emerging role of KDM5A in human cancer, J. Hematol. Oncol. 14 (2021) 30. [144] Y.-F. Wang, J. Zhang, Y. Su, Y.-Y. Shen, D.-X. Jiang, Y.-Y. Hou, M.-Y. Geng,
[122] J. Sterling, S.V. Menezes, R.H. Abbassi, L. Munoz, Histone lysine demethylases J. Ding, Y. Chen, G9a regulates breast cancer growth by modulating iron
and their functions in cancer, Int. J. Canc. 148 (2021) 2375e2388. homeostasis through the repression of ferroxidase hephaestin, Nat. Com-
[123] T.E. McAllister, K.S. England, R.J. Hopkinson, P.E. Brennan, A. Kawamura, mun. 8 (2017) 274.
C.J. Schofield, Recent progress in histone demethylase inhibitors, J. Med. [145] K.T. Hua, M.Y. Wang, M.W. Chen, L.H. Wei, C.K. Chen, C.H. Ko, Y.M. Jeng,
Chem. 59 (2016) 1308e1329. P.L. Sung, Y.H. Jan, M. Hsiao, M.L. Kuo, M.L. Yen, The H3K9 methyltransferase
[124] M. Wissmann, N. Yin, J.M. Müller, H. Greschik, B.D. Fodor, T. Jenuwein, G9a is a marker of aggressive ovarian cancer that promotes peritoneal
C. Vogler, R. Schneider, T. Günther, R. Buettner, E. Metzger, R. Schüle, metastasis, Mol. Canc. 13 (2014) 189.
Cooperative demethylation by JMJD2C and LSD1 promotes androgen [146] L. Wei, D.K.C. Chiu, F.H.C. Tsang, C.T. Law, C.L.H. Cheng, S.L.K. Au, J.M.F. Lee,
receptor-dependent gene expression, Nat. Cell Biol. 9 (2007) 347e353. C.C.L. Wong, I.O.L. Ng, C.M. Wong, Histone methyltransferase G9a promotes
[125] S. Hamada, T. Suzuki, K. Mino, K. Koseki, F. Oehme, I. Flamme, H. Ozasa, liver cancer development by epigenetic silencing of tumor suppressor gene
Y. Itoh, D. Ogasawara, H. Komaarashi, A. Kato, H. Tsumoto, H. Nakagawa, RARRES3, J. Hepatol. 67 (2017) 758e769.
M. Hasegawa, R. Sasaki, T. Mizukami, N. Miyata, Design, synthesis, enzyme- [147] L. Hu, M.D. Zang, H.X. Wang, B.G. Zhang, Z.Q. Wang, Z.Y. Fan, H. Wu, J.-F. Li,
inhibitory activity, and effect on human cancer cells of a novel series of L.P. Su, M. Yan, Z.Q. Zhu, Q.M. Yang, Q. Huang, B.Y. Liu, Z.G. Zhu, G9A pro-
jumonji domain-containing protein 2 histone demethylase inhibitors, J. Med. motes gastric cancer metastasis by upregulating ITGB3 in a SET domain-
Chem. 53 (2010) 5629e5638. independent manner, Cell Death Dis. 9 (2018) 278.
[126] Y. Yu, K. Schleich, B. Yue, S. Ji, P. Lohneis, K. Kemper, M.R. Silvis, N. Qutob, [148] X. Zhong, X. Chen, X. Guan, H. Zhang, Y. Ma, S. Zhang, E. Wang, L. Zhang,
E. van Rooijen, M. Werner-Klein, L. Li, D. Dhawan, S. Meierjohann, Y. Han, Overexpression of G9a and MCM7 in oesophageal squamous cell
M. Reimann, A. Elkahloun, S. Treitschke, B. Do €rken, C. Speck, F.A. Mallette, carcinoma is associated with poor prognosis, Histopathology 66 (2015)
L.I. Zon, S.L. Holmen, D.S. Peeper, Y. Samuels, C.A. Schmitt, S. Lee, Targeting 192e200.
the senescence-overriding cooperative activity of structurally unrelated [149] C. Segovia, E. San Jose -Eneriz, E. Munera-Maravilla, M. Martínez-Ferna ndez,
H3K9 demethylases in melanoma, Canc. Cell 33 (2018) 322e336. L. Garate, E. Miranda, A. Vilas-Zornoza, I. Lodewijk, C. Rubio, C. Segrelles,
[127] E.C. Kauffman, B.D. Robinson, M.J. Downes, L.G. Powell, M.M. Lee, D.S. Scherr, L.V. Valc arcel, O. Rabal, N. Casares, A. Bernardini, C. Suarez-Cabrera,
L.J. Gudas, N.P. Mongan, Role of androgen receptor and associated lysine- F.F. Lo pez-Caldero n, P. Fortes, J.A. Casado, M. Duen ~ as, F. Villacampa,
demethylase coregulators, LSD1 and JMJD2A, in localized and advanced J.J. Lasarte, F. Guerrero-Ramos, G. de Velasco, J. Oyarzabal, D. Castellano,
human bladder cancer, Mol. Carcinogen. 50 (2011) 931e944. X. Agirre, F. Pro sper, J.M. Paramio, Inhibition of a G9a/DNMT network trig-
[128] K.H.M. Mak, Y.M. Lam, R.K. Ng, Histone demethylase JMJD2B/KDM4B regu- gers immune-mediated bladder cancer regression, Nat. Med. 25 (2019)
lates transcriptional program via distinctive epigenetic targets and protein 1073e1081.
interactors for the maintenance of trophoblast stem cells, Sci. Rep. 11 (2021) [150] F. Casciello, F. Al-Ejeh, M. Miranda, G. Kelly, E. Baxter, K. Windloch,
884. F. Gannon, J.S. Lee, G9a-mediated repression of CDH10 in hypoxia enhances
[129] Q. Li, L. Shi, B. Gui, W. Yu, J. Wang, D. Zhang, X. Han, Z. Yao, Y. Shang, Binding breast tumour cell motility and associates with poor survival outcome,
of the JmjC demethylase JARID1B to LSD1/NuRD suppresses angiogenesis Theranostics 10 (2020) 4515e4529.
and metastasis in breast cancer cells by repressing chemokine CCL14, Canc. [151] C.J. Bergin, A. Zouggar, J.R. Haebe, A.N. Masibag, F.M. Desrochers, S.Y. Reilley,
Res. 71 (2011) 6899e6908. G. Agrawal, Y.D. Benoit, G9a controls pluripotent-like identity and tumor-
[130] W. Zhang, J. Cheng, P. Diao, D. Wang, W. Zhang, H. Jiang, Y. Wang, Thera- initiating function in human colorectal cancer, Oncogene 40 (2021)
peutically targeting head and neck squamous cell carcinoma through syn- 1191e1202.
ergistic inhibition of LSD1 and JMJD3 by TCP and GSK-J1, Br. J. Canc. 122 [152] H. Cao, L. Li, D. Yang, L. Zeng, Y. Xie, B. Yu, G. Liao, J. Chen, Recent progress in
(2019) 528e538. histone methyltransferase (G9a) inhibitors as anticancer agents, Eur. J. Med.
[131] D. Rotili, S. Tomassi, M. Conte, R. Benedetti, M. Tortorici, G. Ciossani, Chem. 179 (2019) 537e546.
S. Valente, B. Marrocco, D. Labella, E. Novellino, A. Mattevi, L. Altucci, [153] S. Jan, M.I. Dar, R. Wani, J. Sandey, I. Mushtaq, S. Lateef, S.H. Syed, Targeting
A. Tumber, C. Yapp, O.N. King, R.J. Hopkinson, A. Kawamura, C.J. Schofield, EHMT2/G9a for cancer therapy: progress and perspective, Eur. J. Pharmacol.
A. Mai, Pan-histone demethylase inhibitors simultaneously targeting 893 (2021) 173827.
Jumonji C and lysine-specific demethylases display high anticancer activ- [154] M.R.C. Charles, A. Dhayalan, H.-P. Hsieh, M.S. Coumar, Insights for the design
ities, J. Med. Chem. 57 (2014) 42e55. of protein lysine methyltransferase G9a inhibitors, Future Med. Chem. 11
[132] R. Benedetti, C. Dell'Aversana, T. De Marchi, D. Rotili, N.Q. Liu, B. Novakovic, (2019) 993e1014.
S. Boccella, S. Di Maro, S. Cosconati, A. Baldi, E. Nimeus, J. Schultz, U. Ho
€glund, [155] S. Kubicek, R.J. O'Sullivan, E.M. August, E.R. Hickey, Q. Zhang, Miguel
S. Maione, C. Papulino, U. Chianese, F. Iovino, A. Federico, A. Mai, L. Teodoro, S. Rea, K. Mechtler, J.A. Kowalski, C.A. Homon, T.A. Kelly,
H.G. Stunnenberg, A. Nebbioso, L. Altucci, Inhibition of histone demethylases T. Jenuwein, Reversal of H3K9me2 by a small-molecule inhibitor for the G9a
LSD1 and UTX regulates ERa signaling in breast cancer, Cancers 11 (2019) histone methyltransferase, Mol. Cell. 25 (2007) 473e481.

25
Y.-C. Duan, S.-J. Zhang, X.-J. Shi et al. European Journal of Medicinal Chemistry 222 (2021) 113588

[156] M. Kuppuswamy, S. Vijayalingam, L.J. Zhao, Y. Zhou, T. Subramanian, [176] G. Lopez, Y. Song, R. Lam, D. Ruder, C.J. Creighton, H.K. Bid, K.L. Bill,
J. Ryerse, G. Chinnadurai, Role of the PLDLS-binding cleft region of CtBP1 in S. Bolshakov, X. Zhang, D. Lev, R.E. Pollock, HDAC inhibition for the treatment
recruitment of core and auxiliary components of the corepressor complex, of epithelioid sarcoma: novel cross talk between epigenetic components,
Mol. Cell Biol. 28 (2008) 269e281. Mol. Canc. Res. 14 (2015) 35e43.
[157] Y. Shi, J.i. Sawada, G. Sui, E.B. Affar, J.R. Whetstine, F. Lan, H. Ogawa, M. Po- [177] J.K. Lue, S.A. Prabhu, Y. Liu, Y. Gonzalez, A. Verma, P.S. Mundi, N. Abshiru,
Shan Luke, Y. Nakatani, Y. Shi, Coordinated histone modifications mediated J.M. Camarillo, S. Mehta, E.I. Chen, C. Qiao, R. Nandakumar, S. Cremers,
by a CtBP co-repressor complex, Nature 422 (2003) 735e738. N.L. Kelleher, O. Elemento, J.E. Amengual, Precision targeting with EZH2 and
[158] I. Gyory, J. Wu, G. Fejer, E. Seto, K.L. Wright, PRDI-BF1 recruits the histone H3 HDAC inhibitors in epigenetically dysregulated lymphomas, Clin. Canc. Res.
methyltransferase G9a in transcriptional silencing, Nat. Immunol. 5 (2004) 25 (2019) 5271e5283.
299e308. [178] Y. Wang, S.Y. Chen, S. Colborne, G. Lambert, C.Y. Shin, N.D. Santos,
[159] Z. Duan, A. Zarebski, D. Montoya-Durango, H.L. Grimes, M. Horwitz, Gfi1 K.A. Orlando, J.D. Lang, W.P.D. Hendricks, M.B. Bally, A.N. Karnezis, R. Hass,
coordinates epigenetic repression of p21Cip/WAF1 by recruitment of histone T.M. Underhill, G.B. Morin, J.M. Trent, B.E. Weissman, D.G. Huntsman, His-
lysine methyltransferase G9a and histone deacetylase 1, Mol. Cell Biol. 25 tone deacetylase inhibitors synergize with catalytic inhibitors of EZH2 to
(2005) 10338e10351. exhibit antitumor activity in small cell carcinoma of the ovary, hypercalce-
[160] X. Liang, J. Zang, X. Li, S. Tang, M. Huang, M. Geng, C.J. Chou, C. Li, Y. Cao, mic type, Mol. Canc. Therapeut. 17 (2018) 2767e2779.
W. Xu, H. Liu, Y. Zhang, Discovery of novel Janus kinase (JAK) and histone [179] N. Grinshtein, C. Rioseco, R. Marcellus, D. Uehling, A. Aman, X. Lun, O. Muto,
deacetylase (HDAC) dual inhibitors for the treatment of hematological ma- L. Podmore, G. Cairncross, S. Robbins, S. Jones, M. Marra, R. Al-awar,
lignancies, J. Med. Chem. 62 (2019) 3898e3923. D. Senger, D. Kaplan, Small molecule epigenetic screen identifies novel EZH2
[161] Y. Hu, Y. Zheng, M. Dai, J. Wu, B. Yu, H. Zhang, W. Kong, H. Wu, X. Yu, Snail2 and HDAC inhibitors that target glioblastoma brain tumor-initiating cells,
induced E-cadherin suppression and metastasis in lung carcinoma facilitated Neuro Oncol. 18 (2016) vi75evi76.
by G9a and HDACs, Cell Adhes. Migrat. 13 (2019) 284e291. [180] G. Stazi, L. Taglieri, A. Nicolai, A. Romanelli, R. Fioravanti, S. Morrone,
[162] H.J. Son, J.Y. Kim, Y. Hahn, S.B. Seo, Negative regulation of JAK2 by H3K9 M. Sabatino, R. Ragno, S. Taurone, M. Nebbioso, R. Carletti, M. Artico,
methyltransferase G9a in leukemia, Mol. Cell Biol. 32 (2012) 3681e3694. S. Valente, S. Scarpa, A. Mai, Dissecting the role of novel EZH2 inhibitors in
[163] K.-B. Kim, H.-J. Son, S. Choi, J.Y. Hahm, H. Jung, H.J. Baek, H. Kook, Y. Hahn, primary glioblastoma cell cultures: effects on proliferation, epithelial-
H. Kook, S.-B. Seo, H3K9 methyltransferase G9a negatively regulates UHRF1 mesenchymal transition, migration, and on the pro-inflammatory pheno-
transcription during leukemia cell differentiation, Nucleic Acids Res. 43 type, Clin. Epigenet. 11 (2019) 173.
(2015) 3509e3523. [181] A. Romanelli, G. Stazi, R. Fioravanti, C. Zwergel, E. Di Bello, S. Pomella,
[164] L. Zang, S.M. Kondengaden, Q. Zhang, X. Li, D.K. Sigalapalli, C. Perrone, C. Battistelli, R. Strippoli, M. Tripodi, D. del Bufalo, R. Rota,
S.M. Kondengadan, K. Huang, K. Kathy Li, S. Li, Z. Xiao, L. Wen, H. Zhu, D. Trisciuoglio, A. Mai, S. Valente, Design of first-in-class dual EZH2/HDAC
B.N. Babu, L. Wang, F. Che, P. George Wang, Structure based design, synthesis inhibitor: biochemical activity and biological evaluation in cancer cells, ACS
and activity studies of small hybrid molecules as HDAC and G9a dual in- Med. Chem. Lett. 11 (2020) 977e983.
hibitors, Oncotarget 8 (2017) 63187e63207. [182] Valentina Speranzini, Dante Rotili, Giuseppe Ciossani, Simona Pilotto,
[165] R. Margueron, D. Reinberg, The Polycomb complex PRC2 and its mark in life, Biagina Marrocco, Mariantonietta Forgione, Alessia Lucidi, Federico Forneris,
Nature 469 (2011) 343e349. Mehdipour Parinaz, Sameer Velankar, A. Antonello Mai, Mattevi, Polymyxins
[166] A. He, X. Shen, Q. Ma, J. Cao, A. von Gise, P. Zhou, G. Wang, V.E. Marquez, and quinazolines are LSD1/KDM1A inhibitors with unusual structural fea-
S.H. Orkin, W.T. Pu, PRC2 directly methylates GATA4 and represses its tures, Sci. Adv. 2 (2016) e1601017.
transcriptional activity, Gene Dev. 26 (2012) 37e42. [183] Z.X. He, B.F. Wei, X. Zhang, Y.P. Gong, L.Y. Ma, W. Zhao, Current development
[167] B. Shi, C. Behrens, V. Vaghani, E.M. Riquelme, J. Rodriguez-Canales, H. Kadara, of CBP/p300 inhibitors in the last decade, Eur. J. Med. Chem. 209 (2021)
H. Lin, J. Lee, H. Liu, I. Wistuba, G. Simon, Oncogenic enhancer of zeste ho- 112861.
molog 2 is an actionable target in patients with non-small cell lung cancer, [184] T. Wu, Y.F. Kamikawa, M.E. Donohoe, Brd4's bromodomains mediate histone
Canc. Med. 8 (2019) 6383e6392. H3 acetylation and chromatin remodeling in pluripotent cells through P300
[168] T. Fukumoto, P.H. Park, S. Wu, N. Fatkhutdinov, S. Karakashev, T. Nacarelli, and Brg1, Cell Rep. 25 (2018) 1756e1771.
A.V. Kossenkov, D.W. Speicher, S. Jean, L. Zhang, T.-L. Wang, I.-M. Shih, [185] C.D. Morrison-Smith, T.M. Knox, I. Filic, K.M. Soroko, B.K. Eschle,
J.R. Conejo-Garcia, B.G. Bitler, R. Zhang, Repurposing pan-HDAC inhibitors for M.K. Wilkens, P.C. Gokhale, F. Giles, A. Griffin, B. Brown, G.I. Shapiro,
ARID1A-mutated ovarian cancer, Cell Rep. 22 (2018) 3393e3400. B.E. Zucconi, P.A. Cole, M.E. Lemieux, C.A. French, Combined targeting of the
[169] K.H. Kim, C.W.M. Roberts, Targeting EZH2 in cancer, Nat. Med. 22 (2016) BRD4eNUTep300 Axis in NUT midline carcinoma by dual selective bro-
128e134. modomain inhibitor, NEO2734, Mol. Canc. Therapeut. 19 (2020) 1406e1414.
[170] R. Duan, W. Du, W. Guo, EZH2: a novel target for cancer treatment, [186] F. Giles, M. Witcher, B. Brown, NEO2734: a novel potent oral dual BET and
J. Hematol. Oncol. 13 (2020) 104. P300/CBP inhibitor, Ann. Oncol. 29 (2018) viii140eviii141.
[171] N. Gulati, W. Be guelin, L. Giulino-Roth, Enhancer of zeste homolog 2 (EZH2) [187] F. Spriano, E. Gaudio, L. Cascione, C. Tarantelli, F. Melle, G. Motta, V. Priebe,
inhibitors, Leuk. Lymphoma 59 (2018) 1574e1585. A. Rinaldi, G. Golino, A.A. Mensah, L. Aresu, E. Zucca, S. Pileri, M. Witcher,
[172] H. He, X. Hu, F. Teng, Z. Liu, Q. Zhang, Z. Feng, Q. Feng, L. Yu, Design and B. Brown, C. Wahlestedt, F. Giles, A. Stathis, F. Bertoni, Antitumor activity of
synthesis of (E)-1,2-diphenylethene-based EZH2 inhibitors, Bioorg. Med. the dual BET and CBP/EP300 inhibitor NEO2734, Blood Adv. 4 (2020)
Chem. Lett 30 (2020) 126957. 4124e4135.
[173] J. van der Vlag, A.P. Otte, Transcriptional repression mediated by the human [188] K.R. Ryan, F. Giles, G.J. Morgan, Targeting both BET and CBP/EP300 proteins
polycomb-group protein EED involves histone deacetylation, Nat. Genet. 23 with the novel dual inhibitors NEO2734 and NEO1132 leads to anti-tumor
(1999) 474e477. activity in multiple myeloma, Eur. J. Haematol. 106 (2020) 90e99.
[174] W. Fiskus, M. Pranpat, M. Balasis, B. Herger, R. Rao, A. Chinnaiyan, P. Atadja, [189] J. Zhou, X. Jiang, S. He, H. Jiang, F. Feng, W. Liu, W. Qu, H. Sun, Rational design
K. Bhalla, Histone deacetylase inhibitors deplete enhancer of zeste 2 and of multitarget-directed ligands: strategies and emerging paradigms, J. Med.
associated polycomb repressive complex 2 proteins in human acute leuke- Chem. 62 (2019) 8881e8914.
mia cells, Mol. Canc. Therapeut. 5 (2006) 3096e3104. [190] X.H. Makhoba, C. Viegas Jr., R.A. Mosa, F.P.D. Viegas, O.J. Pooe, Potential
[175] T. Takashina, I. Kinoshita, J. Kikuchi, Y. Shimizu, J. Sakakibara-Konishi, impact of the multi-target drug approach in the treatment of some complex
S. Oizumi, M. Nishimura, H. Dosaka-Akita, Combined inhibition of EZH2 and diseases, Drug Des. Dev. Ther. 14 (2020) 3235e3249.
histone deacetylases as a potential epigenetic therapy for non-small-cell [191] A.R. de Lera, A. Ganesan, Two-hit wonders: the expanding universe of
lung cancer cells, Canc. Sci. 107 (2016) 955e962. multitargeting epigenetic agents, Curr. Opin. Chem. Biol. 57 (2020) 135e154.

26

You might also like